Transparent Peer Review
Download the complete Review Process [PDF] including:
- reviews
- authors' reply
- editorial decisions
Download the complete Review Process [PDF] including:
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
GENERAL COMMENTS
We thank the three reviewers for their comments on the paper.
We are pleased to see that they consider it be a comprehensive and well-executed study, which clearly establishes a previously overlooked connection between MRTF-SRF signalling and proliferation, and that its conclusions require no further experimentation.
As review 3 points out, this work has implications for cancer biology, and suggests new research routes to understand the relation between cell adhesion, proliferation, and transformation.
However, two referees raise significant concerns about its impact
Review 1 suggests that the paper lacks impact without exploration the wider biological significance of our observations, although it considers it to be a good basic cell biology study. It suggests further work extending the findings to tissue- or tumor-based systems. While we consider such studies worthwhile – indeed we are currently pursuing these directions – we consider them beyond the scope of the present paper.
Review 2 questions the novelty of our findings. We strongly disagree. This is is the first study to show that MRTF-SRF signalling is required for the proliferation of both primary and immortalised fibroblasts, and epithelial cells. We show that MRTF inactivation leads cells to enter a quiescence-like state under conditions that would permit efficient cell cycle progression in wildtype cells. The study will alter the field's perspective on the role of MRTF-SRF signalling, previously viewed as concerned with cell adhesion, morphology, and motility.
Responses to individual reviews (italic) follow in regular text.
RESPONSE TO INDIVIDUAL REVIEWS (comments in italic, response in regular, changes made)
__Reviewer #1 __
*(Evidence, reproducibility and clarity (Required)): *
*The manuscript by Neilsen et al. presents a thorough and well-structured study showing that Myocardin-related transcription factors (MRTF-A/B), via MRTF-SRF, are essential for the proliferation of both primary and immortalized fibroblasts and epithelial cells. Using a combination of knockouts/rescue experiments, cytoskeletal analysis, and transcriptomics, the authors demonstrate that MRTF-SRF signalling controls actin dynamics and contractility-key drivers of cell cycle progression. Notably, they show that the proliferative arrest caused by MRTF loss is reversible, distinguishing it from classical senescence. **
Major points*
Suggestions While the authors argue convincingly against classical senescence, elevated SA-βGal and SASP expression suggest a more nuanced arrest state. It not really clear what this state is or is not, therefore a deeper discussion of possible hybrid or intermediate states would be helpful - maybe potential additional experiments to include or exclude potential explanations - e.g. how does it differ from G0 exit?* Our findings show that MRTF inactivation inhibits cell proliferation under conditions that would permit efficient cell cycle progression in wildtype cells, inducing a state with some features associated with classical senescence, and others conventionally associated with reversible cell cycle arrest/quiescence. The reviewer correctly points out that this raises problems with accurately defining the nature of the MRTF-null proliferation defect.
To our knowledge there are no rigorously defined unambiguous markers for senescence, quiescence, or G0. Indeed, recent studies have shown that senescence and quiescence / G0 states are not as distinct as previously assumed (Anwar et al, 2018; Ashraf et al 2023) as we reviewed in detail in Discussion p27, §2; p28 §3. We therefore do not consider it a productive endeavour to define markers for the MRTF-null state as opposed to defining its mechanistic basis. However, we agree that we should have been clearer about how the phenotypes we observe relate to classical cell arrest states.
We have therefore revised the presentation of the Results to make it clear which features of the non-proliferative state associated with MRTF inactivation are seen in classical senescence, and which are found in reversible cell cycle exit or quiescence.
Things done:
Note that p27 is associated with reversible arrest included on p20§2 line 460. We also explicitly summarised the features of the phenotype at the start of the Discussion.
Sentences added p27§1 lines 626-631.
Emphasis that p27 protein upregulation is associated with reversible cell cycle inhibition and quiescence is added on p28 line 668-669.
The transcriptomic data are strong, but the paper would benefit from zooming in on specific MRTF-SRF targets (e.g., actin isoforms, adhesion molecules) that directly link cytoskeletal regulation to cell cycle control.*
We have now clarified presentation of the RNAseq data in Figure 5 and the data summary tables. Figure 5B now identifies which of those genes showing deficits in MRTF-null MEFs were previously identified as direct genomic targets for MRTF-SRF, and that the majority are cytoskeletal.
A critical question remains as to whether these effects a reflect limitation in one MRTF target gene or several, and how this defect relates to proliferation.
Concerning specific MRTF-SRF gene targets:
Cells lacking cytoplasmic actins are reported to exhibit defective proliferation, (__now noted in Results p23 lines 529-532). __We are currently evaluating whether this defect has similarities with the MRTF-null proliferation phenotype (see Discussion p31, §2).
Previous findings suggest that defective cytoplasmic actin expression may underlie most MRTF knockout phenotypes (Salvany et al, 2014; Maurice et al., 2024) previously noted in the Discussion (see p31, §2).
The myoferlin gene promotes growth of liver cancer cells by inhibiting ERK activation and oncogene induced senescence. We showed that myoferlin expression does not promote proliferation of MRTF-null MEFs in the original submission (see Figure S5E). Additionally, we now point out that the RNAseq data show that myoferlin expression is not significantly affected in MRTF-null MEFs __(new text p23, lines 532-534). __
We interpret this comment as indicating that our paper does not address the wider biological implications of our findings by extension to studies in tissue or tumour systems.
As outlined in our response to review 3, our study provides strong evidence that MRTF-SRF will be required for cell proliferation in settings where physical progression through cell cycle transitions requires high contractility, either owing to intrinsic factors or external physical constraints such as tissue stiffness, fibrosis, or tumour microenvironment.
Discussion now explicitly addresses potential roles for tissue stiffness (pp30§2 lines 717-718, and p32§1 725-727). However, we feel that resolution of this question is beyond the scope of the present paper.
We find it difficult to understand the precise points being made here.
However, transformation has long been known to bypass physical constraints on proliferation such as the requirement for adhesion. Moreover, MRTF-SRF activity is not necessarily required for proliferation of all transformed cells (Hampl et al, 2013; Medjkane et al, 2009; our unpublished data). The relation of our findings to transformation is thus an open question, which we are actively pursuing. Now noted in revised Discussion p32, lines 752-755.
MRTF-independent proliferation of tumor cells could reflect oncogenic signals substituting for MRTF-dependent ones (eg from focal adhesions), or from relief of cytoskeletal contraints on proliferation (adhesion independent proliferation). In contrast, in proliferation of DLC1-deleted cancer cells is dependent on suppression of oncogene-induced senescence by MRTF-SRF signalling (Hampl et al, 2013). These points were already made in Discussion p28, pp30-31.
Although our current work is focussed on cell transformation, we would respectfully suggest the in-depth resolution of this complex question is beyond the scope of the present paper.
See also response to (3) above.
*Reviewer #1 (Significance (Required)): *
*Overall *
This is a well-executed and insightful study that deepens our understanding of how cytoskeletal signals drive proliferation through MRTF-SRF. It broadens the role of this pathway beyond motility and offers new perspectives on mechanotransduction and cellular plasticity. If is weak in its demonstration of biological significance, but if the aim to to present a pure basic cell biology story it is good.
The vast majority of work with the SRF system has led to the common perception that its role is exclusively with cell motility and adhesive processes, not proliferation. The results presented in the paper, even if limited to cell culture models, are therefore novel.
Reviewer #2
(Evidence, reproducibility and clarity (Required)):
*In this manuscript, Nielsen and colleagues examine the impact of MRTF-A/B and SRF gene inactivation on cell proliferation. They performed an extensive body of work (using multiple cell types and multiple clones) to show that MRTF inactivation causes cell cycle arrest and senescence (mimicking the phenotype of SRF knockout cells) although the changes in the expression of various CDK inhibitors were cell-type specific. *
*Very interestingly, simultaneous inactivation of all three major CDK inhibitors failed to rescue MRTF knockout cells from their proliferation defect. Expectedly, MRTF knockout cells exhibited defects in actin cytoskeleton, adhesion, and contractility. Interestingly, hyperactivating Rho also failed to rescue MRTF knockout cells from proliferation defect. The main conclusion of the paper was derived from experiments which showed that inhibition of either ROCK or myosin caused wild-type cells to behave like MRTF knockout cells rather than demonstration of any molecular perturbation that could reverse the proliferation defect of MRTF knockout cells. *
While the experimental studies are thorough and rigorous, a vast majority of the core findings related to the loss-of-function of MRTF that are reported herein (i.e. defects in cell proliferation, elevation of CDK inhibitors, migration, actin cytoskeleton, contractility) are not conceptually new and have been previously reported in other cell systems by several investigators including this research group.
This is the first study showing that MRTF-SRF signalling is required for the proliferation of both primary and immortalised fibroblasts, and epithelial cells. We show that the MRTF-SRF non-proliferative state combines features of both classical senescence and reversible cell cycle exit / quiescence.
The vast majority of previous work with the SRF system has led to the common perception that its role is exclusively related to cell motility and adhesive processes and not proliferation (see Olson and Nordheim 2010). Where proliferation has been examined directly, both others and our own previous studies of the MRTFs in immune cells and cancer cells lines have revealed no direct role in proliferation (Schratt et al, 2001;Medjkane et al 2009; Maurice et al, 2024).
The results presented here are therefore novel.
In the reviewer's opinion, since the authors have not been able to identify a molecular strategy to reverse the proliferation phenotype of MRTF knockout cells, the underlying mechanisms of MRTF-dependent regulation of cell proliferation remain largely unanswered.
Indeed, our attempts to rescue the phenotype (knockouts of the CKIs, and overexpression of different downregulated factors) did not restore proliferation. We therefore now aim to attack the problem (i) through overexpression screens, and (ii) by identifying differences between MRTF-SRF dependent and -independent (eg transformed) cells. However, these are new projects that are beyond the scope of a revised paper.
Other comments: Majority of the immunoblot data have not been quantified.
P16 data in Fig 1G vs Fig S1A are not similar (although the authors mention that the findings are similar)
We have addressed these issues by reorganisation and quantification the immunoblotting data as follows:
Figure 1I moved from former Figure S1A, to replace former Figure 1G. New legend now includes quantitation, and reference to Spearman correlations, p44 lines 834-841.
New Figure S1A displays data from multiple independent experiments with all 3 Mrtfab-/- pools. New legend, p44 lines 997-1002.
Figure S1B legend notes correlation between relative marker expression in untreated WT and Mrtfab-/- cells, p44, lines 1005-1008.
Results text rewritten p17 lines 383-391; no reference to “similar”.
*Reviewer #2 (Significance (Required)): *
*This study aims to investigate a fundamental biological question of how an actin-regulated transcription machinery regulates cell proliferation and is therefore of broad significance. Strengths and limitations of this study are described above. *
Reviewer #3
*(Evidence, reproducibility and clarity (Required)): *
Summary
*The manuscript by Nielsen et al. (Treisman lab) entitled "MRTF-dependent cytoskeletal dynamics drive efficient cell cycle progression" investigates the effects on cell proliferation elicited upon cellular depletion of the transcription factors MRTF-A and MRTF-B. The MRTFs are actin-dependent co-factors of SRF, which direct the transcription of SRF target genes. The MRTF-SRF regulatory circuit defines both the functioning and the control of actin-driven cytoskeletal dynamics. *
*The work presented identifies essential molecular links that interconnect cytoskeleton-dependent cellular activities (cell-cell adhesion, cell-substrate contact, cell spreading) and cell proliferation. *
*General assessment on used methodology. *
*The presented comprehensive body of work is performed competently; it includes all relevant and necessary state-of-the-art technologies. *
Reviewer #3 (Significance (Required)):
Advance
Previously published evidence by others (including the Treisman group) had indicated that SRF does not seem essential for the proliferation of some cell types (i. e., embryonic (stem) cells, activation-dependent immune cells, etc.). In regard to this, the authors discuss in the current manuscript: "Although further work is needed to elucidate the basis for these context-dependent dfferences, our data show that MRTF-SRF signalling is likely to play a more general role in proliferation than previously thought." The current manuscript already delineates this "general role": MRTF-SRF signalling impinges on cell proliferation whenever proliferative activities are dependent upon cytoskeletal dynamics.
We of course support the view that it is MRTF-SRF's role in cytoskeletal dynamics, especially contractility, that is a limiting factor for cell cycle progression in our cells; however, this may not be the cases or other cell types or settings, such adhesion-independent or transformed cells, and/or stiff tissue environments.
We have stated this view more strongly, modifying the abstract and discussion, and rewording the sentence quoted above.
The major point is that MRTF-SRF-dependent proliferation may be more common than previously thought, the field having focussed on its role in cytoskeletal dynamics rather than proliferation.
Abstract lines 48-49; Discussion p28, line 668-669; pp30-31, lines 713-714, 725-727. See also last para pp31/32, __added lines 752-755. __
*The work has implications for cancer biology. It offers new directions to investigate the regulation of proliferative activities of anchorage-independent tumor cells. **
Audience *
*The insights generated serve the wide interests of a large and diverse group of cell and tumor biologists. *
*Reviewers field of expertise (keywords). *
Cytoskeletal dynamics, transcriptional con*
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary
The manuscript by Nielsen et al. (Treisman lab) entitled "MRTF-dependent cytoskeletal dynamics drive efficient cell cycle progression" investigates the effects on cell proliferation elicited upon cellular depletion of the transcription factors MRTF-A and MRTF-B. The MRTFs are actin-dependent co-factors of SRF, which direct the transcription of SRF target genes. The MRTF-SRF regulatory circuit defines both the functioning and the control of actin-driven cytoskeletal dynamics. The work presented identifies essential molecular links that interconnect cytoskeleton-dependent cellular activities (cell-cell adhesion, cell-substrate contact, cell spreading) and cell proliferation.
General assessment on used methodology.
The presented comprehensive body of work is performed competently; it includes all relevant and necessary state-of-the-art technologies.
Advance
Previously published evidence by others (including the Treisman group) had indicated that SRF does not seem essential for the proliferation of some cell types (i. e., embryonic (stem) cells, activation-dependent immune cells, etc.). In regard to this, the authors discuss in the current manuscript: "Although further work is needed to elucidate the basis for these context-dependent dfferences, our data show that MRTFSRF signalling is likely to play a more general role in proliferation than previously thought." The current manuscript already delineates this "general role": MRTF-SRF signalling impinges on cell proliferation whenever proliferative activities are dependent upon cytoskeletal dynamics.
The work has implications for cancer biology. It offers new directions to investigate the regulation of proliferative activities of anchorage-independent tumor cells.
Audience
The insights generated serve the wide interests of a large and diverse group of cell and tumor biologists.
Reviewers field of expertise (keywords).
Cytoskeletal dynamics, transcriptional control.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this manuscript, Nielsen and colleagues examine the impact of MRTF-A/B and SRF gene inactivation on cell proliferation. They performed an extensive body of work (using multiple cell types and multiple clones) to show that MRTF inactivation causes cell cycle arrest and senescence (mimicking the phenotype of SRF knockout cells) although the changes in the expression of various CDK inhibitors were cell-type specific. Very interestingly, simultaneous inactivation of all three major CDK inhibitors failed to rescue MRTF knockout cells from their proliferation defect. Expectedly, MRTF knockout cells exhibited defects in actin cytoskeleton, adhesion, and contractility. Interestingly, hyperactivating Rho also failed to rescue MRTF knockout cells from proliferation defect. The main conclusion of the paper was derived from experiments which showed that inhibition of either ROCK or myosin caused wild-type cells to behave like MRTF knockout cells rather than demonstration of any molecular perturbation that could reverse the proliferation defect of MRTF knockout cells. While the experimental studies are thorough and rigorous, a vast majority of the core findings related to the loss-of-function of MRTF that are reported herein (i.e. defects in cell proliferation, elevation of CDK inhibitors, migration, actin cytoskeleton, contractility) are not conceptually new and have been previously reported in other cell systems by several investigators including this research group. In the reviewer's opinion, since the authors have not been able to identify a molecular strategy to reverse the proliferation phenotype of MRTF knockout cells, the underlying mechanisms of MRTF-dependent regulation of cell proliferation remain largely unanswered.
Other comments: Majority of the immunoblot data have not been quantified. P16 data in Fig 1G vs Fig S1A are not similar (although the authors mention that the findings are similar)
This study aims to investigate a fundamental biological question of how an actin-regulated transcription machinery regulates cell proliferation and is therefore of broad significance. Strengths and limitations of this study are described above.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript by Neilsen et al. presents a thorough and well-structured study showing that Myocardin-related transcription factors (MRTF-A/B), via MRTF-SRF, are essential for the proliferation of both primary and immortalized fibroblasts and epithelial cells. Using a combination of knockouts/rescue experiments, cytoskeletal analysis, and transcriptomics, the authors demonstrate that MRTF-SRF signalling controls actin dynamics and contractility-key drivers of cell cycle progression. Notably, they show that the proliferative arrest caused by MRTF loss is reversible, distinguishing it from classical senescence.
Major points
Suggestions
Overall
This is a well-executed and insightful study that deepens our understanding of how cytoskeletal signals drive proliferation through MRTF-SRF. It broadens the role of this pathway beyond motility and offers new perspectives on mechanotransduction and cellular plasticity. If is weak in its demonstration of biological significance, but if the aim to to present a pure basic cell biology story it is good.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Proposed revision plan
Based on the below reviews, we propose the following revision plan. Briefly:
Reviewer #1
Evidence, reproducibility and clarity
In their manuscript entitled "The synovial lining macrophage layer develops in the first weeks of life in a CSF1- and TGFβ-dependent but monocyte-independent process," the authors explore the developmental trajectory of synovial lining macrophages. They demonstrate that the formation of this specialized macrophage layer is age-dependent and governed by a distinct developmental program that proceeds independently of circulating monocytes. Through scRNA-Seq, the authors show that synovial lining macrophages originate locally from Aqp1⁺ macrophages and are marked by the expression of Csf1r, Tgfbr, and Piezo1. Notably, genetic ablation of each of these factors impaired the development of lining macrophages to varying degrees, suggesting differential contributions of CSF1, TGFβ, and PIEZO1 signaling pathways to their maturation and maintenance.
The manuscript is well written, and the data quality and representation is of a high standard. The authors have employed a sophisticated array of state-of-the-art mouse models and cutting-edge technologies to elucidate the developmental origin of synovial lining macrophages. Notably, the supporting scRNA-Seq datasets are of excellence and provide valuable insights that will likely be of significant interest to researchers in the field of immunology and joint biology. Accordingly, the experimental approach and interpretations regarding macrophage origin are well-founded and compelling. However, in the eye of the reviewer, the section addressing the underlying molecular mechanisms is a bit less convincing. This part of the study appears slightly underdeveloped, and some of the mechanistic claims lack sufficient experimental clarity. A more rigorous experimental investigation would be essential to reinforce the manuscript's conclusions, particularly concerning the data related to Tgfbr and Piezo1, where the current evidence appears insufficiently substantiated.
We thank the reviewer for their positive and constructive evaluation of our manuscript. We agree with them (and the other reviewers) that our functional data on the involvement of TGFβ signaling and mechanical loading/mechanosensing are comparably less convincing and substantiated than our developmental data. We are very grateful for their (and the other reviewers’) suggestions to provide more support for the involvement of these factors in lining macrophage development. However, we think that carrying this out to the same high standard will require substantial time and other resources. We have therefore decided to uncouple this from the developmental data and pursue this in follow-up work. We will re-focus the current manuscript on the developmental data. We have proposed to the editors to instead include additional data on synovial fibroblast development, to complement our macrophage data and also delineate the maturation of their niche, thereby providing a conclusive developmental atlas.
Major point:
The numbers of VSIG4⁺ macrophages appear either unaffected or only minimally altered in both Csf1rMerCreMer Tgfbr2floxed and Fcgr1Cre Piezo1floxed mouse models, respectively. This raises an important question: was the gene deletion efficiency sufficient in each model? Accordingly, the authors are encouraged to include quantitative data on gene deletion efficiency for both mouse models, as this information is critical for interpreting the observed phenotypic outcomes and validating the conclusions regarding gene function. Furthermore, to better assess the impact of Tgfbr2 and Piezo1 disruption, the authors should provide more comprehensive flow cytometry analyses and histological data for these mouse models. Given the apparent homogeneity of VSIG4⁺ macrophages (as shown by the authors themselves), bulk RNA-Seq of sorted Tgfbr2- and Piezo1-deficient VSIG4⁺ macrophages (or from TGFβ-treated animals) would offer valuable insights into both the effectiveness of gene deletion and the molecular pathways governed by TGFβ and PIEZO1 in lining macrophages.
As outlined above, we have decided to uncouple our functional data on TGFβ, Piezo1 and mechanical loading. The points raised here are all very valid, and we will implement your suggestions in our follow-up functional work focusing on signaling events regulating lining macrophage development. On the suggestion to perform bulk RNA sequencing for VSIG4+ macrophages: This is a good one in principle – although we will not be able to use this strategy where we want to assess the consequences of experimental treatments or genetic models on lining macrophage maturation, because acquisition of VSIG4 is a key maturation event that might be impaired in these conditions.
Minor points:
Consistent usage of Cx3cr1-GFP+ nomenclature (for instance: Fig. S1 legend "adult mouse synovial tissue, showing PDGFRα⁺ fibroblasts (yellow) and CX3CR1-GFP⁺ cells (cyan)." versus Fig. 1 legend "Automated spot detection highlights Cx3cr1-GFP⁺ macrophages)".
We will implement these changes.
Unclear Fig. 3 legend: "Representative immunofluorescence images of synovial tissue from Clec9aCre:Rosa26lsl-tdT mice at 3 weeks and in adulthood, showing and tdTomato (yellow) and stained for DAPI (blue), VSIG4 (cyan)" Check 'showing and tdTomato.'
We will implement these changes.
For greater clarity, it would have been helpful if the transcript names had been directly included within Figures 3C, S3A, and S3C.
We will implement these changes.
Page 24: "(Mki67CreERT2:Rosa26lsl-tdT)" Last bracket not superscript.
We will implement these changes.
Page 25: "we again leveraged our scRNAsequencing dataset" Missing punctuation.
We will implement these changes.
Page 27: Fig. 5C legend: " of synovial tissue of 1 week-old, 3 weeks-old and adult mice." Please specify and change to 'adult Csf1rΔFIRE/ΔFIRE mice'.
We will implement these changes.
Page 30: The outcome observed in the Acta1-rtTA:tetO-Cre:ChR2-V5fl mouse model appears to be inconclusive: "This approach resulted in an increased density of VSIG4+ and total (F4/80+) macrophages in the exposed leg of some 5 days-old pups, but others showed the opposite trend (Figure S5D)." This variability may reflect low efficiency of the model or other technical limitations (e.g. muscle contractions frequency or time point of analysis). Given this ambiguity, it is worth reconsidering whether the data are sufficiently robust to warrant inclusion. Should the authors choose to include these findings, further experimentation of appropriate depth and precision is required to allow a conclusive interpretation (either it increases the density of VSIG4+ macrophages or not). The same applies to the Yoda1-treated mice, for which additional data are needed to determine whether VSIG4⁺ macrophage density is truly affected.
We have decided to remove the data on the optogenetic mouse model and Yoda1 treatment and follow-on separately, implementing these suggestions, including proof of concept data for optogenetically induced muscle contractions.
Significance
General assessment: provide a summary of the strengths and limitations of the study. What are the strongest and most important aspects? What aspects of the study should be improved or could be developed? This is a well-designed study that uses cutting-edge methodologies to investigate the developmental trajectory of synovial lining macrophages under homeostatic conditions. The authors present robust experimental evidence and compelling interpretations concerning synovial macrophage origin, which are both well-substantiated and impactful. Nonetheless, from the reviewer's perspective, the section exploring the molecular mechanisms underlying macrophage differentiation is comparatively less convincing. This section appears somewhat underdeveloped, as some of the mechanistic claims lack sufficient depth and experimental rigor to fully substantiate the conclusions.
Describe the nature and significance of the advance (e.g. conceptual, technical, clinical) for the field: In contrast to earlier studies (PMID: 31391580, 32601335), the inclusion of fate-mapping experiments adds an important dimension, offering novel insight into the ontogeny of synovial macrophages. This expanded perspective may prove particularly valuable in advancing our understanding of joint immunology, especially regarding the local origins and lineage relationships of macrophage populations.
Furthermore, the authors present novel insights into the molecular pathways underlying the differentiation and development of synovial lining macrophages. By demonstrating previously unrecognized regulatory mechanisms, this work significantly deepens our understanding of the cellular and transcriptional programs that drive macrophage specialization within the joint microenvironment.
Place the work in the context of the existing literature (provide references, where appropriate): This study builds upon previous work characterizing the macrophage compartment in the joint (PMID: 31391580, 32601335), yet provides a substantially more comprehensive dataset that spans multiple developmental time points and data on the origin of this specialized macrophage subset.
State what audience might be interested in and influenced by the reported findings: Immunologist, clinicians
Define your field of expertise with a few keywords to help the authors contextualize your point of view. Indicate if there are any parts of the paper that you do not have sufficient expertise to evaluate. This study falls well within the scope of the reviewer's expertise in innate immunity.
Reviewer #2
Evidence, reproducibility and clarity
In the manuscript „The synovial lining macrophage layer develops in the first weeks of life in a CSF1- and TGFβ- dependent but monocyte-independent process", Magalhaes Pinto and colleagues carefully employ a wide range of technologies including single cell profiling, imaging and an exceptional combination of fate mapping models to characterize the ontogeny and development of lining macrophages in the joint, thus dissecting their maturation during postnatal development. Over the last decade, several landmark studies highlighted the imprinting of tissue-resident macrophages by a combination of ontogenetic and tissue-specific niche factors during development. So far, the ontogeny and the tissue niche factors governing the development and maturation of lining macrophages have not been described. Therefore, the results of this study offers insights on a small highly adapted macrophage population with relevance in many disease settings in the joint. Furthermore, the findings are nicely showcasing how macrophages are specializing to even very small tissue niches across development within one bigger anatomical compartment to serve dedicated functions within this niche.
This manuscript is beautifully written and highlights many novel, highly relevant findings on lining macrophage biology and the authors employ a wide range of different technologies to carefully dissect the postnatal development of lining macrophages.
In particular, the combination of scRNA-seq and fate mapping is providing a unique the link of transcriptional programs to ontogeny within the tissue niche. Furthermore, the integrative use of distinct fate mapping strategies, transgenic mouse lines, and treatment paradigms to elucidate key niche factors guiding the development and maturation of lining macrophages provides many interesting findings and data that are highly relevant to the field. I really enjoyed reading this manuscript.
Thank you for your complimentary and constructive assessment of our manuscript, and the detailed comments below, which are very helpful. Please find point-by-point responses below.
Major points:
The authors show dynamic regulation of VSIG4 in lining macrophages during development, therefore VSIG4 is maybe not an ideal choice for gating strategies to define lining macrophages or to show as a single markers in immunofluorescence (IF) stainings to demonstrate their abundance across development (even though it is clear that this is the reason why the F4/80 staining is shown next to it). To demonstrate the increase of lining macrophages during development in IF, it would be more helpful if the authors would show quantifications of all F4/80+ cells and additionally VSIG4+ as a proportion of F4/80+ cells (or VSIG4+ F4/80+ and all F4/80+ in a stacked bar plot). We agree with the assessment of VSIG4 not being ideal since this is a key marker of mature lining macrophages only.
We will provide these additional analyses.
In Figure 1C, the authors nicely demonstrate that the lining macrophages get closer in their distance across development to build the epithelial-like macrophage structure along the adult lining. Is the close proximity between lining macrophages already fully "matured" at 3 weeks of age and comparable to adults? Please quantify the distance in adult linings.
We will provide data for adult joints.
Can the authors explain how the grouping was performed between the analyzed human fetal joints? It is not clear why the cut was chosen between the groups at 16/17 weeks of age. Maybe it would be also beneficial if the authors would consider not grouping these samples but rather show the specific quantifications for each samples individually and estimate via linear regression the expansion over time across human development. Furthermore, can the authors give additional information about the distancing of lining macrophages in the human fetal samples, it would be great to see if they follow the same dynamics as in mouse. Maybe comparison to human juvenile/adult joints would also add on to substantiate the findings in human samples (if possible).
We will show samples ungrouped and perform linear regression analysis as suggested.
The scRNA-seq analysis leaves several questions open and some conclusions and workflows cannot be easily followed.
We appreciate this comment and the complexity of the data, and will implement the below recommendations, and clarify the issues raised.
It is not clear how and especially why the signature genes to define macrophages vs. monocytes were chosen. Especially as the signature genes for monocytes would not include patrolling monocytes and the macrophage signature genes seem to be highly regulated during development, see also Apoe expression in NB vs. adult in Figure S2e. Why did the authors not take classical markers such as Itgam, Fcgr1a, Csf1r?
Can dendritic cell signatures be excluded? Cluster 11 and 12 show indeed some DC markers, are these really macrophages?
The authors provide several figure panels showing TOP marker genes or key marker genes for the identified clusters, however it is not clear if these are TOP DE genes or if the genes were hand chosen. Somehow, the authors give the impression that the clusters were chosen and labeled not based on DE genes, but more on existing literature that previously reported these macrophage populations. DE gene lists for all annotated cell types and macrophage clusters need to be provided within the manuscript.
The authors claim that Clusters 1 and 4 are "developing" macrophages. How is this defined? Why are these developing cells compared to other clusters? And why are these clusters later on not considered as progenitors of Aqp1 macrophages and Vsig4 macrophages? Why are Aqp1+ macrophages not labeled as developing when they are later on in the manuscript shown as potential intermediate progenitors of lining macrophages?
Furthermore, it is again confusing that markers are used throughout Figure 2 which are labeled as "key marker genes" for a population and then later on they are claimed to be regulated during development within this population, see for example Figure 2D and 2H.
It is appreciated that the authors distinguished cycling clusters such as 8, 9, and 10 based on their cycling gene signature. Here it would be very exciting to see a cell cycle analysis across all clusters and time points to see when exactly the cells are expanding during development; this would also substantiate the data later shown for the Mki67-CreERT2 mouse model.
Can the authors identify certain gene modules during development of lining macrophages (and/or their progenitors) which are associated with certain functions (e.g. GO terms, GSEA enrichment)?
To determine the actual presence of the identified macrophage clusters from the scRNA-seq as macrophage populations in the joint, the authors should perform IF or FACS for key markers. Especially, Aqp1+ macrophages should be shown in the developing joint.
We will provide additional data, but would also like to reference a study by collaborators currently in revision at Immunity, which characterizes the Aqp1+ population in detail. We are hoping to have a doi available during our revision process.
The authors used a wide range of fate mapping models, which is quite unique and highly appreciated. The obtained results and the conclusions made from the models raise a couple of questions: Whereas contribution of HSC-derived/monocyte-derived macrophages to the lining compartment seems to be minor, there is still labeling across different models. Various aspects would need to be clarified.
We will clarify these data throughout as per below suggestions.
For example, the authors employ Ms4a3-Cre as a tracing model for GMP-derived monocytes, however all quantifications of the labeling efficiency are not normalized to the labeling in monocytes or another highly recombined cell population. This should be shown, similar to the other fate mapping models (Figure 3 F-I).
Labelling efficacy for Ms4a3-Cre is near complete for GMP-derived monocytes (and neutrophils) with the Rosa-lsl-tdT (aka Ai14) reporter we have used (see also PMID: 31491389 and doi: 10.1101/2024.12.03.626330); but we will include normalized data as requested.
Please show Ms4a3 expression across clusters across time points, to exclude expression in fetal-derived clusters.
We will include this in the revised supplementary information, but there is indeed very little at birth (in line with the original report for other tissues PMID: 31491389).
In line with the question raised above, if the authors can exclude a development of the Egfr1+ and Clec4n+ developing macrophages into Aqp1+ macrophages and subsequently into Vsig4 lining macrophages, the obtained data from the Ms4a3-Cre model highly suggests a correlative labeling across these clusters what could implicate a relation. However, the authors do not discuss throughout the manuscript the role of these developing macrophages. It is highly encouraged to include this into the manuscript and it would be of high relevance to understand lining macrophage development.
This is an interesting point and we agree it deserves consideration in the revised manuscript. Indeed, our trajectory analyses do not predict differentiation of the Egfr1+ and Clec4n+ developing macrophages into Aqp1+ macrophages, and hence, ultimately lining macrophages. Conversely, Aqp1+ cells might also convert into Egfr1+ and Clec4n+ developing macrophages. We will elaborate on this more in the revised manuscript.
The authors conclude from the pseudo bulk transcriptomic profiling of the different macrophage clusters that TdT+ and TdT- macrophages do not differ in their gene expression profile and that this is due to niche imprinting rather than origin imprinting. Even though the data supports that conclusion, the authors should verify if inkling cells early during development also show this similar gene expression profile and gene expression should be compared at the different developmental time points. Tissue niche imprinting is happening within the niche during development, most likely in a stepwise progress, and therefore there should be differences in the beginning.
This is another important point that we will address in the revised manuscript by performing additional differential gene expression analyses at the different developmental time points, including the earliest stages, as suggested.
The trajectorial analysis using different pseudotime pipelines is very interesting and nicely points out the potential role of Aqp1 macrophages as intermediates of Vsig4 lining macrophages. From my point of view, all trajectories seem to suggest that Egfr1 developing macrophages and Clec4n developing macrophages might differentiate into Aqp1 macrophages, however the authors are not exploring this further and the role of both developing macrophage clusters is not further discussed (see also comments above).
We will address and discuss this in the revised manuscript.
How was the starting point of the trajectorial analyses defined and is it the same for each pipeline used?
We will clarify this in the revised manuscript.
Are there potentially two trajectories? It looks like there is one in the beginning of postnatal life and a second one appearing from the monocyte-compartment later in life. If this is true, that would rather speak for a dual ontogeny of Vsig4+ macrophages, wouldn't it?
We will discuss this in the revised manuscript.
A heatmap (transcriptional shift) of trajectories between more clusters should be shown at least for Cluster 0,1,2, and 3. It is not sufficient to demonstrate this only between two clusters.
We will add these analyses during revision.
To show the similarity between Aqp1 macrophages and proliferating macrophage clusters, the authors should remove the cycling signature and compare these clusters to show that the cycling cells might be Aqp1 macrophages or earlier developing macrophage progenitors aka Clec4n or Egfr1 macrophages.
We will address this in the revised manuscript.
The conclusions made from the Mki67-CreERT2 data are a bit difficult to understand, whereas all progenitors (monocyte progenitors and macrophage progenitors will proliferate at the neonatal time point and no conclusions can be made if the cells expand in the niche. The authors should employ Confetti mice or other models (Ubow mice) to analyze clonal expansion in the niche.
We agree that interpretation of the Mki67-CreERT2 data is complicated by labeling of other cells, and notably, labeling observed in BM-derived cells. We will highlight this better in the revised manuscript. We have tried using Ubow mice to address this issue, but the recombination efficacy we yielded was too low to draw conclusions. We will address this during revision.
All predicted cell-cell interactions between macrophages and fibroblasts should be provided in a supplementary table. Are the interactions shown in Figure 5 chosen interactions or the TOP predicted ones? Whereas the authors show different numbers of interactions, it is most likely hand-picked and therefore biased.
We will provide a full list of all predicted interactions in the revised supplementary material in addition to a list of the full differential gene expression analysis.
The authors further aim to dissect the factors involved in the developmental niche imprinting of lining macrophages. Even though it is highly appreciated that the authors used so many experimental setups to show the reliance of lining macrophages on Csf1 and TGF-beta as well as mechanosensation, the wide range of models the different methods used and selected developmental time points make it very difficult to really interpret the data. The authors should carefully choose time points and methods (either FACS analysis across all models or IF across all, or both). Often deletion efficiencies for transgenic models and proof of concept that the inhibitors and agonists are working in the treatment paradigm are not provided. For example, Csf1rMer-iCre-Mer Tgfbr2fl/fl mice are used but no deletion efficiency is shown or different time points of analysis, maybe the macrophages are not properly targeted in the set up.
We have decided to uncouple our experimental data on Tgfb, Piezo1 and mechanosensing/mechanical loading, but are taking this into consideration for revision. In many cases, we have in fact performed flow cytometry and imaging analyses, and agree, we should be showing this consistently.
The authors have shown the role of Csf1 and Tgfbr2 only for lining macrophages, is this specific in the joint to this population of are subliming macrophages affected in a similar manner.
We will include data on sublining macrophages in the revised figure (for CSF1; Tgfb data will be uncoupled from this current manuscript).
Can the authors confirm their results in CSF1R-FIRE mice with anti-Csf1 injections or in Csf1op/op mice?
We will expand our discussion of the Csf1 findings, and will consider including anti-CSF1 data during revision. Phenotypes on other Csf1(r) deficient mice are published, if not with the same developmental resolution as our time course in Csf1rFIRE knockout mice and with simpler readouts. Csf1op/op mice are indeed deficient in synovial lining macrophages, from 2 days of age onwards (PMID: 8050349), and lining macrophages are also absent from 2-weeks-old and adult Csf1r-/- mice (PMID: 11756160).
The setup in Figure S5G is very interesting to test the role of movement and mechanical load on the joint, however, there is basically no data on the model provided showing the efficiency of the induced optogenetic muscle contractions, and only one time point is shown.
Data on mechanical loading will be uncoupled from the current manuscript and substantiated in a separate follow-up.
The results regarding the role of Piezo1 and mechanosensation vary a lot. Could it be that analyses were done too early or that actually proper weight load on the joint must be applied for the maturation of the macrophages? The authors should test this to.
We will uncouple these data from the current manuscript during revision. However, this is a possibility that we have discussed. In fact, the most appropriate experimental approach to address the involvement of mechanical loading, onset of walking and specifically, weight bearing would be a loss-of-function approach (i.e. paralysis at the newborn stage), for which we unfortunately could not obtain ethics approval from the UK Home Office.
The Rolipram experiment is shown in Figure S5G, but is not described in the result section. It only appears at some point in the discussion part. The authors should move it to results or remove it from the manuscript.
We will incorporate these data with the revised section on developing synovial macrophage populations.
Minor points:
Please reference the Figure panels in numeric order throughout the text.
We will change this where not the case.
Figure 2a and 2b are a bit out of the storyline, it is not obvious why this is shown here and maybe it would be good to move it to the supplements. Gating strategy is also not used for scRNA-seq. Therefore, it would better fit to the later analysis of joint macrophages across different transgenic mouse models and treatment paradigms. The gating strategies are changing across different experiments throughout the figures, it would be nice to have a similar gating strategy for all experiments, see also Figure 3 where the defining markers for joint macrophages are changing between models.
We will revise Figures 2, 3 and the related supplementary figures.
A lot of figure panels have very small labeling that is basically unreadable. Axes at FACS plots for example. Sometimes, it is even impossible to distinguish cluster labels especially when they have similar colors.
We will revise this, thanks for pointing it out.
In the text on page 14, many markers are named which are specifically regulated during development in lining macrophages, but these factors are not labeled anywhere in the volcano plot. It would be good to showcase at least some of these named genes in the figure panel, e.g. Trem2.
We will do this for revision.
Figure 2F and Figure S2F are really nicely showing the percentage of cells per cluster in each analyzed biological sample. Maybe the authors could additionally consider to show a stacked bar plot with the mean percentage of cells per cluster and how the clusters are distributed across time points?
We will include this in the revised manuscript.
Figure 3A: IF for adult lining macrophages and the quantification are missing.
This will be included in the revised version.
Significance
This manuscript highlights novel, highly relevant findings on lining macrophage biology and the authors employ a wide range of different technologies to carefully dissect the postnatal development of lining macrophages. Furthermore, this study showcases in a very elegant and detailed way the adaptation of macrophage progenitors to a highly specific anatomical tissue niche.
The manuscript is of high interest to basic scientists focussing on macrophage biology and immune cell development and clinicians and clinician scientists focussing on joint diseases such as RA.
Therefore the manuscript is of interest to a wide community working in immunology.
Reviewer #3
Summary:
Magalhaes Pinto, Malengier-Devlies, and co-authors investigated the developmental origins and maturation of synovial (lining and sublining) macrophages across embryonic, newborn, and postnatal stages in mouse. The authors used multiple transgenic reporter lines, lineage tracing, scRNA-seq, 2D confocal and 3D lightsheet imaging, and perturbations to delineate the macrophage states and ontogeny. They propose a model in which the majority of the joint lining macrophages has a fetal (EMP-derived) origin and a small proportion has a definitive HSC-derived monocyte origin, which both seed and mature within the synovial space in the postnatal period in the first 3 weeks of life. Using cell-cell communication analysis on their scRNA-seq data, they identified Fgf2, Csf1, and Tgfb as candidate signaling pathways that support (lining) macrophage development and maturation. Functional experiments indicate that the process is CSF1 and TGFb-dependent and also partly dependent on mechanosensing through Piezo1.
The key conclusions on the composition of the synovial macrophages are convincing based on the presented results, and are carefully phrased. The study is very comprehensive, yet the description and organization of the results of the different mouse models could be altered to improve the storyline. Several refinements in data presentation, formulation, and minor validation experiments would further improve the clarity of the story, as well as summary recaps of the major findings throughout the text.
We thank this reviewer for their detailed review. We will be implementing the requested changes wherever technically feasible.
Major comments:
Generally, the story could be more streamlined by introducing earlier reporter lines and lineage-origin logic. Clearly state which reporter/CreERT2 lines and acrosses are used. It was unclear in Figure 2 that cells of the cross of the Cx3cr1-GFP and Ms4a3Cre:Rosa26lsl-tdT reporter lines were used for the scRNA-seq. The principle that there are fetal-derived and bone marrow (GMP)-derived monocytes and macrophages doesn't need to be "hidden" until Figure 3. For example, also the imaging of Ms4a3Cre could be introduced before the scRNA-seq.
We will revise the structure and order of the manuscript during revision.
Figure 1 could benefit from a cartoon visualizing the anatomy of the knee joint. The terms "sublining" and "synovium" are now a bit unclear, as it appears that sometimes the synovium is indicated as sublining and vice versa. Additionally, a schematic developmental timeline could be added to indicate the parallels between mouse and human development (fetal and postnatal development in mouse versus gestational age in human). Also, the various waves of hematopoiesis could be indicated in this timeline, which would be particularly helpful for Figure 3 for the lineage-tracing readouts. Lastly, the authors could end the manuscript (a new Figure 6) with a general cartoon summarizing all the results presented.
We will include illustrations as suggested.
Figure 1 could be rearranged: first introduce the markers CX3CR1 and VSIG4 (Figure 1D) and then present the quantifications (Figure 1B/E). Where possible, co-visualization CX3CR1-GFP and VSIG4 on tissue sections to strengthen the claims on the relationship between these 2 markers. Tying the scRNA-seq insights (Figure 2) to the imaging would be elegant. Moreover, it would be informative to represent the CX3CR1+ and VSIG4+ macrophages as a percentage of F4/80+ macrophages (Figure 1B/E). Similarly, for the flow cytometry data in Figure 2, the relationship between the markers CX3CR1 and VSIG4 on macrophages could be more clearly displayed and discussed.
Thanks for this remark. We will endeavor to show co-localization and analysis of both markers wherever possible. However, where we did not use Cx3cr1gfp mice, co-staining was limited by antibody choice.
The 3D imaging of the joint is a nice addition to the manuscript, as it provides more context to the anatomical structure; however, while the text suggests several newborn joints were imaged, Figure 1F visualizes (again) the knee joint. Could other joints also be represented by 3D imaging? If the knee joint is the only joint available for imaging, and previous confocal imaging focused specifically on the meniscus in the knee joint, could the meniscus also be highlighted in the lightsheet imaging?
Apologies if this was not clear from the original manuscript text, but we have only imaged the knee joint in 3D. We will clarify this during revision and consider inclusion of additional imaging data.
Clarification is requested regarding the imaging quantification representation. The M&M section under "Statistical analysis and reproducibility" states that individual data points are displayed, and bars represent the mean. However, some of the Figure legends (e.g., Figures 1B and S1C) specify that each dot corresponds to an individual mouse, with quantification based on 2-3 sections per mouse. While this appears to be a very reasonable representation of the data, does this mean that for each dot, the mean value from the 2-3 sections per mouse was calculated and plotted?
We will clarify this.
It is not clear how the differential expression analysis was performed on the Vsig4+ cells. Please specify if Cluster 0 was used for analysis, or all Vsig4-expressing cells? Not all cells in Cluster 0 have Vsig4+ expression. The authors described the expression dynamics of Aqp1 as intriguing, but lack a reasoning on why this is interesting.
We will revise this section.
Figure S3E: In line with the previous comment, can the authors justify that the tdTomato+/- comparisons are not biased by scRNA-seq dropout (scRNA-seq is zero-inflated, so some tdTomato- cells could be false negatives), and provide methodological details (thresholds, ambient RNA correction, etc.) to support this?
We will clarify this and include additional representations of the tdTomato transcript data.
Although the sex-related differences in macrophage composition and the absence of differential expression are interesting, they distract from the manuscript's main messages. Moreover, the Discussion does not elaborate on how these observations relate to joint (disease) biology. Consider removing this section or integrating it clearly into the relevant biological context.
We will remove this section as suggested.
CreERT2 transgenic lines are often not 100% efficient in recombination, also depending on whether tamoxifen or 4-OHT is used. Could the authors report the percentage of tdTomato+ cells in the joints and compare them to the recombination efficiencies in the monocytes/microglia under the same tamoxifen or 4-OHT conditions? This would help clarify how the interpret the macrophage labeling %'s.
We will report labelling efficacies and/or show normalized data in the revised manuscript.
Could the authors draw parallels between the observations in the mouse knee joint macrophage populations and literature on other joints in mouse and the knee joint in human (for example, as described in Alivernini et al., 2020 and in the very recent Raut et al., 2025)?
We will include a section on this in the revised manuscript.
Minor comments:
In general, the authors should clarify in the Results what each marker used for imaging, flow cytometry, or in the mouse reporter lines delineates. For example, mention that F4/80 is a marker for tissue-resident macrophages (correct?) in immunofluorescence, that IBA1 is a marker for macrophages on human tissue sections (Figure S1), and PDPN is GP38 (Figure S2 - align usage of marker reference across main text and figures).
We will implement this request.
For clarity in the microscopy representation, the single channels should be represented in a grey scale.
We will revise image presentation.
Figure S1B: Is CX3CR1 also restricted to the lining macrophages in human? Could a co-staining with IBA1 be performed to strengthen the species similarities?
To our knowledge, there is no antibody available that works for imaging of human CX3CR1. Moreover, CX3CR1 is only limited to the lining population in adult joints, in fetal and newborn (mouse) joints, all macrophages express this receptor, as do fetal progenitors to macrophages. However, Alivernini and colleagues have reported that TREM2high macrophages are the human counterpart of the mouse CX3CR1+ lining population (PMID: 32601335).
Adipocyte diameter quantification: Avoid plotting individual adipocytes from 2 mice without per-mouse visualization. Instead, report the mean adipocyte diameter per mouse and plot those means.
We will implement this change.
A little typo was spotted in the "Statistical analysis and reproducibility" section: it is Dunn's, not Bunn's multiple-comparison correction.
Thanks for spotting this.
Figure 2A: The gating strategy for the CX3CR1-GFP cells is missing.
We will provide this in the revised manuscript or supplementary material.
Improve the visualization of some plots. For example, Figure 2F is hard to read because of the big dot size. The dots seem to add no information to the graph and could be removed. Additionally, for comparing the clusters across the different time points, one could project the cells from the other time points in grey in the background.
We will revise the presentation of these data.
Figure S2: The dotplot is more informative than the heatmap, consider removing the heatmap.
We will do that.
Figure 3A: If technically feasible, image and visualize both the GFP and tdTomato expression. It would be informative to see the Cx3cr1+ and Ms4a3-derived cells in the same specimen.
We will thrive to show this in the revised manuscript.
Figure 3C: Highlight that tdTomato expression is visualized here.
We will do that.
Figure 3G,F: The authors should place the schematics and graphs next to each other, so the data points can be more easily compared.
We aim to do this in the revised manuscript.
Figure 4B: Which co-staining was performed for the immunofluorescence to quantify the % of tdTomato+ cells?
We co-stained for F4/80 and assessed localization in the lining or sublining. This will be clarified in the revised Figure legend.
Figure 4C: The trajectory analysis appears to have an arrow pointing from the Ccr2+ macrophages to the Ly6c+ monocytes. Please verify this directionality, as its seems against the known biology.
This will be addressed during revision.
Figure 5 mentions that the Csfr1 levels were reduced in a tissue-specific manner, but it is unclear how this tissue specificity was achieved.
We apologize for this misunderstanding. Csfr1FIRE mice are not tissue-specific knockouts, but they are more specific than global knockout mice, since only a (myeloid-specific) enhancer is affected. We will clarify this in the relevant section.
For the TGFb perturbations (Tgfbr2 KO and systemic TGFb depletion): did the authors validate reduced TGFb pathway activity in the macrophages, for example, reduced pSMAD2/3 levels? This would validate the effectiveness of the perturbations. This is an important point, and assessing signaling events downstream of TGFb is a very good suggestion.
As per above comment, we have decided to uncouple the functional data with exception of CSF1 from the revised version of the current manuscript, but we will be taking this into account for substantiating our functional data in follow-up work.
Figure 5F could benefit from a timeline of the treatment.
As for the previous point raised, we will be taking this into account for follow-up work on the uncoupled functional data.
The Methods mention that Gene Ontology analysis was performed on the single-cell data, but the results are not plotted in a figure. It would be informative to include this GO/pathway analysis in the appropriate figure(s).
We will include this in the revised (supplementary) information.
Significance:
This work provides a high temporal-resolution and "spatial" resolution reference map of the ontogeny and maturation of the synovial lining macrophages in the knee joint. It complements existing literature that demonstrated the presence of tissue-resident macrophages in the synovial space and lining (Culemann, et al., 2019 and others) by charting the embryonic-to-postnatal emergence of lining and sublining subsets. In particular, this mouse work identified some key signaling pathways in shaping this tissue compartment. This dataset serves as a robust, steady-state reference for joint pathology and can be implemented with human studies on disease biology of the knee joint (e.g., Alivernini et al., 2020; Raut et al., 2025). Insights into the exact developmental origins, mechanisms contributing to diverse or seemingly similar cell types, and distinct maturation processes are crucial to understanding disease biology, in which developmental processes can be hijacked/reactivated.
These findings will interest researchers in joint disease biology (osteoarthritis and immune-mediated arthritides such as RA and psoriasis), macrophage development (tissue-resident vs monocyte-derived lineages), the bone/joint microenvironment, and joint mechanobiology.
The reviewer's expertise is in developmental biology, mesoderm, bone biology, hematopoiesis, and monocyte/macrophage biology in disease
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
Magalhaes Pinto, Malengier-Devlies, and co-authors investigated the developmental origins and maturation of synovial (lining and sublining) macrophages across embryonic, newborn, and postnatal stages in mouse. The authors used multiple transgenic reporter lines, lineage tracing, scRNA-seq, 2D confocal and 3D lightsheet imaging, and perturbations to delineate the macrophage states and ontogeny. They propose a model in which the majority of the joint lining macrophages has a fetal (EMP-derived) origin and a small proportion has a definitive HSC-derived monocyte origin, which both seed and mature within the synovial space in the postnatal period in the first 3 weeks of life. Using cell-cell communication analysis on their scRNA-seq data, they identified Fgf2, Csf1, and Tgfb as candidate signaling pathways that support (lining) macrophage development and maturation. Functional experiments indicate that the process is CSF1 and TGFb-dependent and also partly dependent on mechanosensing through Piezo1. The key conclusions on the composition of the synovial macrophages are convincing based on the presented results, and are carefully phrased. The study is very comprehensive, yet the description and organization of the results of the different mouse models could be altered to improve the storyline. Several refinements in data presentation, formulation, and minor validation experiments would further improve the clarity of the story, as well as summary recaps of the major findings throughout the text.
Major comments:
Minor comments:
This work provides a high temporal-resolution and "spatial" resolution reference map of the ontogeny and maturation of the synovial lining macrophages in the knee joint. It complements existing literature that demonstrated the presence of tissue-resident macrophages in the synovial space and lining (Culemann, et al., 2019 and others) by charting the embryonic-to-postnatal emergence of lining and sublining subsets. In particular, this mouse work identified some key signaling pathways in shaping this tissue compartment. This dataset serves as a robust, steady-state reference for joint pathology and can be implemented with human studies on disease biology of the knee joint (e.g., Alivernini et al., 2020; Raut et al., 2025). Insights into the exact developmental origins, mechanisms contributing to diverse or seemingly similar cell types, and distinct maturation processes are crucial to understanding disease biology, in which developmental processes can be hijacked/reactivated.
These findings will interest researchers in joint disease biology (osteoarthritis and immune-mediated arthritides such as RA and psoriasis), macrophage development (tissue-resident vs monocyte-derived lineages), the bone/joint microenvironment, and joint mechanobiology.
The reviewer's expertise is in developmental biology, mesoderm, bone biology, hematopoiesis, and monocyte/macrophage biology in disease
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In the manuscript „The synovial lining macrophage layer develops in the first weeks of life in a CSF1- and TGFβ- dependent but monocyte-independent process", Magalhaes Pinto and colleagues carefully employ a wide range of technologies including single cell profiling, imaging and an exceptional combination of fate mapping models to characterize the ontogeny and development of lining macrophages in the joint, thus dissecting their maturation during postnatal development. Over the last decade, several landmark studies highlighted the imprinting of tissue-resident macrophages by a combination of ontogenetic and tissue-specific niche factors during development. So far, the ontogeny and the tissue niche factors governing the development and maturation of lining macrophages have not been described. Therefore, the results of this study offers insights on a small highly adapted macrophage population with relevance in many disease settings in the joint. Furthermore, the findings are nicely showcasing how macrophages are specializing to even very small tissue niches across development within one bigger anatomical compartment to serve dedicated functions within this niche.
This manuscript is beautifully written and highlights many novel, highly relevant findings on lining macrophage biology and the authors employ a wide range of different technologies to carefully dissect the postnatal development of lining macrophages.
In particular, the combination of scRNA-seq and fate mapping is providing a unique the link of transcriptional programs to ontogeny within the tissue niche. Furthermore, the integrative use of distinct fate mapping strategies, transgenic mouse lines, and treatment paradigms to elucidate key niche factors guiding the development and maturation of lining macrophages provides many interesting findings and data that are highly relevant to the field. I really enjoyed reading this manuscript.
Major points:
1) The authors show dynamic regulation of VSIG4 in lining macrophages during development, therefore VSIG4 is maybe not an ideal choice for gating strategies to define lining macrophages or to show as a single markers in immunofluorescence (IF) stainings to demonstrate their abundance across development (even though it is clear that this is the reason why the F4/80 staining is shown next to it). To demonstrate the increase of lining macrophages during development in IF, it would be more helpful if the authors would show quantifications of all F4/80+ cells and additionally VSIG4+ as a proportion of F4/80+ cells (or VSIG4+ F4/80+ and all F4/80+ in a stacked bar plot).
2) In Figure 1C, the authors nicely demonstrate that the lining macrophages get closer in their distance across development to build the epithelial-like macrophage structure along the adult lining. Is the close proximity between lining macrophages already fully "matured" at 3 weeks of age and comparable to adults? Please quantify the distance in adult linings.
3) Can the authors explain how the grouping was performed between the analyzed human fetal joints? It is not clear why the cut was chosen between the groups at 16/17 weeks of age. Maybe it would be also beneficial if the authors would consider not grouping these samples but rather show the specific quantifications for each samples individually and estimate via linear regression the expansion over time across human development. Furthermore, can the authors give additional information about the distancing of lining macrophages in the human fetal samples, it would be great to see if they follow the same dynamics as in mouse. Maybe comparison to human juvenile/adult joints would also add on to substantiate the findings in human samples (if possible).
4) The scRNA-seq analysis leaves several questions open and some conclusions and workflows cannot be easily followed.
a. It is not clear how and especially why the signature genes to define macrophages vs. monocytes were chosen. Especially as the signature genes for monocytes would not include patrolling monocytes and the macrophage signature genes seem to be highly regulated during development, see also Apoe expression in NB vs. adult in Figure S2e. Why did the authors not take classical markers such as Itgam, Fcgr1a, Csf1r?
b. Can dendritic cell signatures be excluded? Cluster 11 and 12 show indeed some DC markers, are these really macrophages?
c. The authors provide several figure panels showing TOP marker genes or key marker genes for the identified clusters, however it is not clear if these are TOP DE genes or if the genes were hand chosen. Somehow, the authors give the impression that the clusters were chosen and labeled not based on DE genes, but more on existing literature that previously reported these macrophage populations. DE gene lists for all annotated cell types and macrophage clusters need to be provided within the manuscript.
d. The authors claim that Clusters 1 and 4 are "developing" macrophages. How is this defined? Why are these developing cells compared to other clusters? And why are these clusters later on not considered as progenitors of Aqp1 macrophages and Vsig4 macrophages? Why are Aqp1+ macrophages not labeled as developing when they are later on in the manuscript shown as potential intermediate progenitors of lining macrophages?
e. Furthermore, it is again confusing that markers are used throughout Figure 2 which are labeled as "key marker genes" for a population and then later on they are claimed to be regulated during development within this population, see for example Figure 2D and 2H.
f. It is appreciated that the authors distinguished cycling clusters such as 8, 9, and 10 based on their cycling gene signature. Here it would be very exciting to see a cell cycle analysis across all clusters and time points to see when exactly the cells are expanding during development; this would also substantiate the data later shown for the Mki67-CreERT2 mouse model.
g. Can the authors identify certain gene modules during development of lining macrophages (and/or their progenitors) which are associated with certain functions (e.g. GO terms, GSEA enrichment)?
5) To determine the actual presence of the identified macrophage clusters from the scRNA-seq as macrophage populations in the joint, the authors should perform IF or FACS for key markers. Especially, Aqp1+ macrophages should be shown in the developing joint.
6) The authors used a wide range of fate mapping models, which is quite unique and highly appreciated. The obtained results and the conclusions made from the models raise a couple of questions: Whereas contribution of HSC-derived/monocyte-derived macrophages to the lining compartment seems to be minor, there is still labeling across different models. Various aspects would need to be clarified.
a. For example, the authors employ Ms4a3-Cre as a tracing model for GMP-derived monocytes, however all quantifications of the labeling efficiency are not normalized to the labeling in monocytes or another highly recombined cell population. This should be shown, similar to the other fate mapping models (Figure 3 F-I).
b. Please show Ms4a3 expression across clusters across time points, to exclude expression in fetal-derived clusters.
c. In line with the question raised above, if the authors can exclude a development of the Egfr1+ and Clec4n+ developing macrophages into Aqp1+ macrophages and subsequently into Vsig4 lining macrophages, the obtained data from the Ms4a3-Cre model highly suggests a correlative labeling across these clusters what could implicate a relation. However, the authors do not discuss throughout the manuscript the role of these developing macrophages. It is highly encouraged to include this into the manuscript and it would be of high relevance to understand lining macrophage development.
d. The authors conclude from the pseudo bulk transcriptomic profiling of the different macrophage clusters that TdT+ and TdT- macrophages do not differ in their gene expression profile and that this is due to niche imprinting rather than origin imprinting. Even though the data supports that conclusion, the authors should verify if inkling cells early during development also show this similar gene expression profile and gene expression should be compared at the different developmental time points. Tissue niche imprinting is happening within the niche during development, most likely in a stepwise progress, and therefore there should be differences in the beginning.
7) The trajectorial analysis using different pseudotime pipelines is very interesting and nicely points out the potential role of Aqp1 macrophages as intermediates of Vsig4 lining macrophages. From my point of view, all trajectories seem to suggest that Egfr1 developing macrophages and Clec4n developing macrophages might differentiate into Aqp1 macrophages, however the authors are not exploring this further and the role of both developing macrophage clusters is not further discussed (see also comments above).
8) How was the starting point of the trajectorial analyses defined and is it the same for each pipeline used?
9) Are there potentially two trajectories? It looks like there is one in the beginning of postnatal life and a second one appearing from the monocyte-compartment later in life. If this is true, that would rather speak for a dual ontogeny of Vsig4+ macrophages, wouldn't it?
10) A heatmap (transcriptional shift) of trajectories between more clusters should be shown at least for Cluster 0,1,2, and 3. It is not sufficient to demonstrate this only between two clusters.
11) To show the similarity between Aqp1 macrophages and proliferating macrophage clusters, the authors should remove the cycling signature and compare these clusters to show that the cycling cells might be Aqp1 macrophages or earlier developing macrophage progenitors aka Clec4n or Egfr1 macrophages.
12) The conclusions made from the Mki67-CreERT2 data are a bit difficult to understand, whereas all progenitors (monocyte progenitors and macrophage progenitors will proliferate at the neonatal time point and no conclusions can be made if the cells expand in the niche. The authors should employ Confetti mice or other models (Ubow mice) to analyze clonal expansion in the niche.
13) All predicted cell-cell interactions between macrophages and fibroblasts should be provided in a supplementary table. Are the interactions shown in Figure 5 chosen interactions or the TOP predicted ones? Whereas the authors show different numbers of interactions, it is most likely hand-picked and therefore biased.
14) The authors further aim to dissect the factors involved in the developmental niche imprinting of lining macrophages. Even though it is highly appreciated that the authors used so many experimental setups to show the reliance of lining macrophages on Csf1 and TGF-beta as well as mechanosensation, the wide range of models the different methods used and selected developmental time points make it very difficult to really interpret the data. The authors should carefully choose time points and methods (either FACS analysis across all models or IF across all, or both). Often deletion efficiencies for transgenic models and proof of concept that the inhibitors and agonists are working in the treatment paradigm are not provided. For example, Csf1rMer-iCre-Mer Tgfbr2fl/fl mice are used but no deletion efficiency is shown or different time points of analysis, maybe the macrophages are not properly targeted in the set up.
15) The authors have shown the role of Csf1 and Tgfbr2 only for lining macrophages, is this specific in the joint to this population of are subliming macrophages affected in a similar manner.
16) Can the authors confirm their results in CSF1R-FIRE mice with anti-Csf1 injections or in Csf1op/op mice?
17) The setup in Figure S5G is very interesting to test the role of movement and mechanical load on the joint, however, there is basically no data on the model provided showing the efficiency of the induced optogenetic muscle contractions, and only one time point is shown.
18) The results regarding the role of Piezo1 and mechanosensation vary a lot. Could it be that analyses were done too early or that actually proper weight load on the joint must be applied for the maturation of the macrophages? The authors should test this to
19) The Rolipram experiment is shown in Figure S5G, but is not described in the result section. It only appears at some point in the discussion part. The authors should move it to results or remove it from the manuscript.
Minor points:
1) Please reference the Figure panels in numeric order throughout the text.
2) Figure 2a and 2b are a bit out of the storyline, it is not obvious why this is shown here and maybe it would be good to move it to the supplements. Gating strategy is also not used for scRNA-seq. Therefore, it would better fit to the later analysis of joint macrophages across different transgenic mouse models and treatment paradigms. The gating strategies are changing across different experiments throughout the figures, it would be nice to have a similar gating strategy for all experiments, see also Figure 3 where the defining markers for joint macrophages are changing between models.
3) A lot of figure panels have very small labeling that is basically unreadable. Axes at FACS plots for example. Sometimes, it is even impossible to distinguish cluster labels especially when they have similar colors.
4) In the text on page 14, many markers are named which are specifically regulated during development in lining macrophages, but these factors are not labeled anywhere in the volcano plot. It would be good to showcase at least some of these named genes in the figure panel, e.g. Trem2.
5) Figure 2F and Figure S2F are really nicely showing the percentage of cells per cluster in each analyzed biological sample. Maybe the authors could additionally consider to show a stacked bar plot with the mean percentage of cells per cluster and how the clusters are distributed across time points?
6) Figure 3A: IF for adult lining macrophages and the quantification are missing
This manuscript highlights novel, highly relevant findings on lining macrophage biology and the authors employ a wide range of different technologies to carefully dissect the postnatal development of lining macrophages. Furthermore, this study showcases in a very elegant and detailed way the adaptation of macrophage progenitors to a highly specific anatomical tissue niche.
The manuscript is of high interest to basic scientists focussing on macrophage biology and immune cell development and clinicians and clinician scientists focussing on joint diseases such as RA
Therefore the manuscript is of interest to a wide community working in immunology.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In their manuscript entitled "The synovial lining macrophage layer develops in the first weeks of life in a CSF1- and TGFβ-dependent but monocyte-independent process," the authors explore the developmental trajectory of synovial lining macrophages. They demonstrate that the formation of this specialized macrophage layer is age-dependent and governed by a distinct developmental program that proceeds independently of circulating monocytes. Through scRNA-Seq, the authors show that synovial lining macrophages originate locally from Aqp1⁺ macrophages and are marked by the expression of Csf1r, Tgfbr, and Piezo1. Notably, genetic ablation of each of these factors impaired the development of lining macrophages to varying degrees, suggesting differential contributions of CSF1, TGFβ, and PIEZO1 signaling pathways to their maturation and maintenance.
The manuscript is well written, and the data quality and representation is of a high standard. The authors have employed a sophisticated array of state-of-the-art mouse models and cutting-edge technologies to elucidate the developmental origin of synovial lining macrophages. Notably, the supporting scRNA-Seq datasets are of excellence and provide valuable insights that will likely be of significant interest to researchers in the field of immunology and joint biology. Accordingly, the experimental approach and interpretations regarding macrophage origin are well-founded and compelling. However, in the eye of the reviewer, the section addressing the underlying molecular mechanisms is a bit less convincing. This part of the study appears slightly underdeveloped, and some of the mechanistic claims lack sufficient experimental clarity. A more rigorous experimental investigation would be essential to reinforce the manuscript's conclusions, particularly concerning the data related to Tgfbr and Piezo1, where the current evidence appears insufficiently substantiated.
Major point:
Minor points:
This is a well-designed study that uses cutting-edge methodologies to investigate the developmental trajectory of synovial lining macrophages under homeostatic conditions. The authors present robust experimental evidence and compelling interpretations concerning synovial macrophage origin, which are both well-substantiated and impactful. Nonetheless, from the reviewer's perspective, the section exploring the molecular mechanisms underlying macrophage differentiation is comparatively less convincing. This section appears somewhat underdeveloped, as some of the mechanistic claims lack sufficient depth and experimental rigor to fully substantiate the conclusions. - Describe the nature and significance of the advance (e.g. conceptual, technical, clinical) for the field:
In contrast to earlier studies (PMID: 31391580, 32601335), the inclusion of fate-mapping experiments adds an important dimension, offering novel insight into the ontogeny of synovial macrophages. This expanded perspective may prove particularly valuable in advancing our understanding of joint immunology, especially regarding the local origins and lineage relationships of macrophage populations. Furthermore, the authors present novel insights into the molecular pathways underlying the differentiation and development of synovial lining macrophages. By demonstrating previously unrecognized regulatory mechanisms, this work significantly deepens our understanding of the cellular and transcriptional programs that drive macrophage specialization within the joint microenvironment. -Place the work in the context of the existing literature (provide references, where appropriate):
This study builds upon previous work characterizing the macrophage compartment in the joint (PMID: 31391580, 32601335), yet provides a substantially more comprehensive dataset that spans multiple developmental time points and data on the origin of this specialized macrophage subset. - State what audience might be interested in and influenced by the reported findings:
Immunologist, clinicians - Define your field of expertise with a few keywords to help the authors contextualize your point of view. Indicate if there are any parts of the paper that you do not have sufficient expertise to evaluate.
This study falls well within the scope of the reviewer's expertise in innate immunity.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
We would like to thank the reviewers for their overall positive evaluations of our manuscript and for their invaluable suggestions that will allow us to reinforce our conclusions. We acknowledge that there is some work to be done and are ready to address most of the reviewers' comments as detailed in our replies below.
Reviewer #1
The findings that mmDicer is proviral in bat cells relies exclusively on the observation that the depletion of Dicer in M. myotis cells leads to a reduced accumulation of SFV and SINV at the RNA and protein levels (figure 2). Heterologous expression of mmDicer in HEK 293T NoDice doesn't lead to an increase permissivity to viral infections (figure 1) and the accumulation of Dicer foci is only observed in M. myotis cells but not when mmDicer is expressed in HEK 293 NoDice cells (figure 6). Given that the key finding of this manuscript relies on these knockdown experiments, the authors should ensure that the impact on viral infections is due to the specific silencing of mmDicer and not caused by off-target effects of their siRNA-mediated approach. The authors designed a siRNA pool to efficiently knock-down mmDicer. They should validate their findings by using individual Dicer siRNA and verify whether the decrease SFV/SINV accumulation is observed with at least two individual siRNAs targeting Dicer. It would also strengthen their findings if they could show a complementation experiment in which a mmDicer (designed to not be affected by the siRNA-mediated silencing) is introduced exogenously in Dicer-depleted cells and show that it rescues the observed decrease in viral accumulation to demonstrate that the proviral role is strictly dependent on mmDicer. Alternatively, the authors could consider a CRISPR/Cas9 genome editing approach to knockout Dicer in bat cells to test whether this proviral effect is confirmed.
Reply: We agree with this reviewer that it is important to provide evidence for the specificity of the knock-down and to rule out any off-target effect of the siRNAs. This is the reason for using the siTool technology, which relies on the use of a pool of 30 siRNAs that are transfected at a final concentration of 3 nM. This means that each individual siRNA in the pool is at a concentration of 0.1 nM, so the possibility of off-target effect is largely avoided and the efficiency of silencing is boosted by the cooperative activity of many siRNAs (see https://www.sitoolsbiotech.com/documents/sipools/siPOOLBrochure2019_Web.pdf for more details). This being said, we agree that it would be better to confirm that the observed effect can be recapitulated using a single siRNA and that a complementation experiment would definitely strengthen our findings. For this reason, we will test two individual siRNAs targeting the 3' UTR of mmDicer, which will allow us to complement the knock-down by transfecting a cDNA construct. Regarding the CRISPR/Cas9 genome editing approach, we will give it a try, but Dicer is notoriously difficult to knock-out, so we cannot be sure that this will be successful.
Figure 2: the authors knock-downed Dicer in M. myotis nasal epithelial cells and carried out infections with SINV-GFP and SFV. The authors conclude that Dicer is proviral as its depletion causes a decrease in SINV-GFP and SFV accumulation. While this conclusion is supported by the decrease levels of viral RNA and protein levels upon Dicer depletion (figure 2D, 2E, 2G), the effect on the viral titers is non-significant for both viruses (Figure 2C and 2F) based on the statistical analysis. This reviewer appreciates that the titers are lower upon Dicer knockdown, which support the authors' findings at the viral RNA and protein levels. However, as these results are central to the core message of the manuscript, the authors should provide evidence that this proviral effect observed is statistically significant on viral titers by perhaps providing additional repeats and/or comment on this observation.
Reply: Indeed, we agree that even if the effect of Dicer knockdown results in a lowering of the viral titer, it would be better to have a statistically significant effect. We will repeat the experiment to increase the number of replicates and the power of the statistical test.
a) *In figure 4 and 5, the authors nicely show that mmDicer accumulate to cytoplasmic foci in M. myotis cells upon infection with SFV and SINV and these foci co-localise with double-stranded RNA. The authors used a commercial polyclonal antibody against Dicer (A301-937A, Bethyl according to the Material and Methods section) which is specific to human Dicer to carry out their immunostaining in bat cells. The authors should provide evidence that this antibody indeed recognises/crossreacts with mmDicer as well and that the staining shown is indeed specific to mmDicer localisation especially because the heterologous expression of HA-tagged version of mmDicer in HEK 293T NoDice cells did not show this accumulation of cytoplasmic foci. The authors should verify the specificity of their mmDicer immunostaining by performing the same labelling in bat cells in which Dicer is knock-downed (or knock out) by individual and validated siRNA against mmDicer. The decrease signal of bat Dicer staining using the anti-human Dicer antibody would indicate specificity. *
Reply: the reviewer is correct in its assertion and it is important to provide evidence that the protein that is detected by the anti-human Dicer antibody in bat cells is indeed Dicer. We will perform the suggested experiment and do an immunostaining using the Dicer antibody in bat cells upon Dicer knockdown.
b) Another complementary approach would be to test their Dicer staining between HEK NoDice cells (no Dicer present) versus NoDice complemented with either mmDicer or human Dicer constructs, which would then indicate how much the anti-human Dicer antibody recognises bat Dicer.
Reply: this complementary approach should yield even cleaner result than the previous one as there will be no expression of Dicer at all in the HEK NoDice cells. Therefore, we should be able to measure the increase of signal in the IF upon expression of either human or bat Dicer. We will perform this experiment together with the other one suggested above. In addition, since the constructs are tagged, we might be able to do a double-staining and verify the colocalization of the two signals.
c) In addition, the authors should overexpress HA-tagged mmDicer in M. myotis nasal epithelial cells and test whether HA-mmDicer accumulate into foci upon infection using an anti-HA immunostaining. This would confirm that these accumulation into foci indeed is specific to mmDicer but also would reinforce the authors' findings that host factors within bat cells are important for this formation into foci since mmDicer expression in HEK 293T No Dice cells didn't show this phenotype upon infection (figure 6). OPTIONAL: it would be interesting to overexpress HA-tagged human Dicer into M. myotis nasal epithelial cells as well to then test using anti-HA staining whether human Dicer in presence of host factors from the bat can accumulate into cytoplasmic foci or not upon viral infection.
Reply: we could perform the suggested experiment, but we might face the issue that transfected cells might mount an immune response, which makes them resistant to the infection. We have observed indeed that we needed to use a higher MOI to infect cells after they have been transfected. Since we will have controls in place, this might not be too much of a problem, but we will have to keep it in mind. Alternatively, we will perform a lentiviral transduction of the cells.
This reviewer appreciates that this might be judged as beyond the scope of this study since it is focused on the role of Dicer in M. myotis. However, the observation that mmDicer accumulates into foci containing as well viral dsRNA is very interesting and it would significantly improve the manuscript if the authors would provide further indications that this phenotype is related to the lack of antiviral activity of mmDicer compared to what has been previously shown in other bat species (P.alecto and T. brasiliensis). In other words, is this accumulation of mmDicer into foci responsible for its different impact on virus infection? It would therefore be insightful to compare Dicer localisation upon infection in M. myotis versus P.alecto and/or T. brasiliensis bat cells in which Dicer was shown to be antiviral and test whether this accumulation in foci is only observed in bat cells in which Dicer is proviral (M. myotis) but not in the other bat cells in which Dicer is antiviral (P.alecto and/or T. brasiliensis).
Reply: this is something that we have been wondering about and we have therefore started to look for the cell lines that have been described in the two published studies. While it proved difficult to find the PaKi cells from P. alecto bats, which is not commercially available, we have obtained the Tblu cells from T. brasiliensis and will look at Dicer localization in this model. However, we have to pay attention to the fact that the published data reported a contribution of RNAi in this cell line upon SARS-CoV-2 infection and that we will be using SINV. In addition, we do not know yet whether the anti-Dicer antibody will cross react with the T. brasiliensis Dicer protein.
OPTIONAL: Given the difference between the provial role of mmDicer compared to the antiviral activity of Dicer in cells from P.alecto and T. brasiliensis bat cells, it would strengthen the authors' findings. if additional experiments would be conducted in parallel using M. myotis, P.alecto and/or T. brasiliensis cells. Notably knocking down Dicer in both M. myotis, P.alecto and/or T. brasiliensis cells, compare the impact on viral infections with SINV, SFV, VSV and correlate any observed difference in phenotype with putative variations in the formation of foci.
Reply: it would indeed be really nice to be able to do the Dicer knockdown experiment in several bat cell lines and to correlate the phenotype with the formation of foci. This experiment might take a long time and we are not sure to be able to realize it in a reasonable amount of time. It could however be the subject of another manuscript further down the line.
*Minor comments *
- Figure 2I: The authors performed a knockdown of Dicer in M. myotis nasal epithelial cells and monitor the impact on VSV-GFP infection. They found that knocking down Dicer leads to an increase in GFP protein and RNA levels suggesting an antiviral role of Dicer while, in contrast, no effect is observed on the production of infectious particles (figure 2H). On the western blot there is only a slight/weak increase of GFP protein level observed upon Dicer knockdown. Yet, the quantification of the band intensity shows a 4-fold increase relative to tubulin and compared to cells treated with siRNA control. This 4-fold increase seems exaggerated given the low increase in the intensity shown on the blot. This discrepancy is most likely due to the lower intensity of tubulin in the western blot analysis of siDicer-treated cells compared to siNeg-treated cells. The authors should reload their western blot with equal amount of protein extract loaded to ensure that the results shown on the western blot are in line with the quantification.*
Reply: the signal quantification for this experiment was done across several replicates, but we agree that the observed effect seems exaggerated when compared to the signal seen on the blot. We observed important variations between replicates, but we will make sure that this was not due to a problem in the analysis and reload the western blot if needed.
- Figure 3D: the authors mention that in both HEK293T cells and M. myotis nasal epithelial cells infected with SINV-GFP, there was an enrichment of 22-nucleotides (nt) paired positive and negative sense reads that overlapped with a 2-nt overhang, typical of Dicer cleavage. In Figure 3D, the data shows indeed that the duplexes are enriched for reads of 22-nt but it is unclear how this analysis reveals a 3' 2nt overhang within these duplexes. Can the authors clarify this point and if the data provided in that particular analysis indeed doesn't allow to detect these overhangs, please rephrase accordingly or provide additional analysis to support that point. *
Reply: In Figure 3D, the graphs show the probability of pairing of all 22 nucleotides sequence mapping either to the plus or the minus strand of the viral RNA. Thus, for each sequence mapping to the plus strand, the number of sequences mapping to the minus strand with a full or partial overall is counted. A corresponding probability of pairing and Z score is calculated for each number of overlapping nucleotides (for more information on the calculation see Antoniewski (2014) Computing siRNA and piRNA Overlap Signatures. In Animal Endo-SiRNAs: Methods and Protocols, Werner A (ed) pp 135-146. New York, NY: Springer). The Z score peaks for an overlap of 20 nt in both HEK293T and M. myotis nasal epithelial cells infected with SINV. This means that there is a higher probability of two 22 nt sequence to pair along 20 nt, and thus that there are two unpaired nucleotides at the extremities of the duplexes. This higher Z score at 20 nt is not seen in VSV-infected cells. We will rephrase the text in the manuscript to make this point clearer.
- Typo: page 5, line 152: the authors mention that Dicer knock down had an antiviral effect against VSV-GFP infection at the RNA and protein levels. However, the data in Figure 2I and 2J show an increase in both GFP RNA and proteins levels upon knockdown of Dicer. Although this data suggests that Dicer is antiviral against VSV, the knockdown of Dicer itself is not antiviral but rather proviral/increase virus accumulation. Please rephrase this sentence to avoid confusions. *
Reply: thank you for spotting this typo. We have corrected it accordingly.
Reviewer #2.
Figure 1 relies on transduction of cells and antibiotic selection to obtain mmDicer-expressing cells. Although we would expect that every cell expresses the construct of interest, this is not always the case, depending on the cell type and toxicity of the construct. As the constructs are tagged, I suggest that the authors use flow cytometry to measure expression levels in a single cell manner. While doing so, they can infect with SINV-GFP and correlate GFP signal with construct expression in each cell, providing a more accurate measurement of mmDicer effect on viral infection. Alternatively, the authors could use live microscopy, as done in Fig 2, to obtain similar data.
Reply: the reviewer is correct that we did not go for monoclonal selection of our mmDicer-expressing cells and therefore that there could be some cell-to-cell variation in expression. However, we have done immunostaining of Dicer in these cells and did not see drastic differences in expression, so we do not think this should impact SINV-GFP expression in a major way. We will provide these images and a quantification of the Dicer signal as a supplementary figure.
For Fig 1C and 1F, it would be great to have growth curves with two different MOIs, instead of a single time point, to ensure that a putative antiviral effect is not missed. Same goes for Fig 2C, especially when the authors document quite a big defect on GFP expression (a proxy for SINV infection) when Dicer is knocked down (Fig 2B). There may be a bigger difference in titers at earlier time points. This matter runs throughout the manuscript. I do not suggest that the authors should provide growth curves every time viral titers are measured, but it is still worth doing it for the 2-3 key experiments of the paper.
Reply: we will perform growth curves of virus infection for the key experiments in the manuscript as suggested. We already have done kinetic measurements of GFP accumulation at different MOIs, which we can provide as supplementary data, but we agree with the reviewer that GFP signal should not been used as the only proxy for the infection and that measuring viral titers by plaque assay is important as well.
Figure 4, could the authors provide a proof that the Dicer antibody is specific in the bat context? This can be done by staining Dicer in bat cells knocked down for Dicer and infected with SINV. The apparition of foci upon anti-Dicer antibody staining should be abbrogated or severely impaired by the knock-down.
Reply: see our reply to point 3 of Reviewer 1.
Fig 5C, please provide a quantification of the images.
Reply: these microscopy images have not been quantified because they have been obtained with an epifluorescence microscope. Indeed, the Pearson correlation coefficient can only be obtained using a confocal microscope. In fact, we have tried to use a confocal microscope to take pictures of these FISH images, but the SINV gRNA signal was too weak or the dots too small to be properly visualized. Furthermore, there is a very large difference in signal intensity between HEK293T and M. myotis cells, making it difficult to define a signal threshold compatible for both cell lines.
l.263, when comparing this work with the recent publications on bat antiviral RNAi, the authors could also provide the percentage identity between Dicers from different species.
Reply: this is a valid point, we have looked at the percentage identity between Dicer proteins from different bat species but we did not include this in our manuscript. We will provide this analysis in the revised version together with a comparison of Dicer from other mammals as a reference point.
Reviewer 3.
- Without direct comparison to the other bat species Dicers (especially where RNAi activity has been suggested as antiviral in previous publications) there is little in this paper that can be concluded about global aspects of bat dicer/RNAi.*
Reply: see our reply to point 4 of Reviewer 1. We are planning to look at least in Tblu cells whether there is also a relocalization of Dicer upon SINV infection. So far, we could not obtain PaKi cells, but we are still looking and should we get those, we will test them as well.
*Minor *
What rules out that the mmDicer re-localization observed in the immortalized mm nasal epithelial is due simply to greater expression levels over the NoDice cells heterologously expressing mmDicer?
Reply: we will provide an immunoblot to show the level of Dicer expression between HEK NoDice + mmDicer and M. myotis nasal epithelial cells as suggested below to address this point.
- Although partially addressed in the text stating the generally long half-life of miRNAs, it seems the simplest explanation for this observation is due to some activity of a shorter-lived miRNA is required for optimal alphavirus replication is the mm nasal epithelial cells. *
Reply: this is an interesting hypothesis that would prove difficult to test in a reasonable amount of time. We thank the reviewer and will mention this possibility in the discussion of the revised manuscript.
*Suggestions that could enhance the magnitude of conclusions that can be drawn from this work. *
*Major *
- Making NoDice cells expressing other bat species Dicers, including those with claims that RNAi is antiviral, would address how universal these current observations are to bats/cell lines.*
Reply: this could be an alternative to the use of P. alecto or T. brasiliensis cell lines that we have mentioned above. We will try to clone Dicer from the Tblu cells that we have in the laboratory. Since we do not have PaKi cells at the moment, it will be more complicated for the Pteropus Dicer, but one possibility could be to synthesize it. However, Dicer is a big gene so it could prove tricky.
- Including an immunoblot showing that mm cells express mmDicer no more abundantly than the heterologous NoDice cells would allow ruling out the trivial explanation that foci occur at a certain critical mass of Dicer*
Reply: yes, we will provide this piece of data as stated in reply to point 2.
*Minor *
- I believe line 151 " In contrast, Dicer * *knock down had an ANTIVIRAL effect against VSV-GFP infection at the RNA and protein *
*levels, but no difference in titers was found (Fig. 2H-J)." should be " In contrast, Dicer *
*knock down had an PROVIRAL effect against VSV-GFP infection at the RNA and protein *
*levels, but no difference in titers was found (Fig. 2H-J)." *
Reply: thank you for spotting this error, which was also mentioned by Reviewer 1, we have corrected this in the text.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this manuscript by Gaucherand and colleagues, the authors demonstrate that heterologous expression of Myotis myotis Dicer into 293 derivative Dicer KO cells did not produce antiviral effects. The authors further demonstrate that knockdown of Dicer in SV40 immortalized M myotis nasal epithelial cells results in reduced alphavirus infection. Finally, they show a correlation where mmDicer changes subcellular localization co-localizing with likely alphavirus replication foci. The manuscript is clearly written, and the conclusions drawn as stated are accurate.
Strengths
Critiques
Major
Suggestions that could enhance the magnitude of conclusions that can be drawn from this work.
Major
As written, this work would be significant to aficionados of bat RNAi. With a little extra work, this could have broader significance regarding more global aspect of Dicer in the the bat antiviral response.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This study by the Pfeffer lab interrogates the role of Dicer during RNA virus infection in bats. This is an interesting and important topic, as bats are well-documented to be a reservoir of viruses that can target humans. The field of bat immunology is gaining momentum, but there is still a lot to be done. This study is thus particularly timely. It also explores more of a niche pathway when it comes to immunity: antiviral RNAi and its entry point, Dicer. This work comes after two recent studies, cited by the authors (Dai 2024, Owolabi 2025), that also explore this concept. Here though, the Pfeffer lab comes to an opposite conclusion, as their results advocate against the existence of antiviral RNAi in bats. As discussed by the authors, discrepancies between their study and the two others may be linked to differences in experimental systems. It nonetheless brings a novel, interesting take on the topic of Dicer & antiviral RNAi in bats, and will be of interest to immunologists and virologists. Altogether, I find the manuscript well-written and clear. Experiments are to the point and well interpreted. Below are a few suggestions that will help bolster the authors' conclusions.
Figure 1 relies on transduction of cells and antibiotic selection to obtain mmDicer-expressing cells. Although we would expect that every cell expresses the construct of interest, this is not always the case, depending on the cell type and toxicity of the construct. As the constructs are tagged, I suggest that the authors use flow cytometry to measure expression levels in a single cell manner. While doing so, they can infect with SINV-GFP and correlate GFP signal with construct expression in each cell, providing a more accurate measurement of mmDicer effect on viral infection. Alternatively, the authors could use live microscopy, as done in Fig 2, to obtain similar data.
For Fig 1C and 1F, it would be great to have growth curves with two different MOIs, instead of a single time point, to ensure that a putative antiviral effect is not missed. Same goes for Fig 2C, especially when the authors document quite a big defect on GFP expression (a proxy for SINV infection) when Dicer is knocked down (Fig 2B). There may be a bigger difference in titers at earlier time points. This matter runs throughout the manuscript. I do not suggest that the authors should provide growth curves every time viral titers are measured, but it is still worth doing it for the 2-3 key experiments of the paper.
Figure 4, could the authors provide a proof that the Dicer antibody is specific in the bat context? This can be done by staining Dicer in bat cells knocked down for Dicer and infected with SINV. The apparition of foci upon anti-Dicer antibody staining should be abbrogated or severely impaired by the knock-down.
Fig 5C, please provide a quantification of the images.
l.263, when comparing this work with the recent publications on bat antiviral RNAi, the authors could also provide the percentage identity between Dicers from different species.
This study by the Pfeffer lab interrogates the role of Dicer during RNA virus infection in bats. This is an interesting and important topic, as bats are well-documented to be a reservoir of viruses that can target humans. The field of bat immunology is gaining momentum, but there is still a lot to be done. This study is thus particularly timely. It also explores more of a niche pathway when it comes to immunity: antiviral RNAi and its entry point, Dicer. This work comes after two recent studies, cited by the authors (Dai 2024, Owolabi 2025), that also explore this concept. Here though, the Pfeffer lab comes to an opposite conclusion, as their results advocate against the existence of antiviral RNAi in bats. As discussed by the authors, discrepancies between their study and the two others may be linked to differences in experimental systems. It nonetheless brings a novel, interesting take on the topic of Dicer & antiviral RNAi in bats, and will be of interest to immunologists and virologists. Altogether, I find the manuscript well-written and clear. Experiments are to the point and well interpreted. Below are a few suggestions that will help bolster the authors' conclusions.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Bats acts a reservoir for many viruses. While some of these viruses can be pathogenic for humans and other animals, infected bats tolerate these viruses and show little to no pathogenesis. It is therefore key to characterise which immune pathways are active in bats and how do they differ from other mammals to understand how bats can sustain these virus infections. RNA interference (RNAi) acts as an antiviral mechanism in plants, invertebrates and was recently shown to be active in a cell type-dependent manner as a defence mechanism in mammals. Notably, recent findings show that antiviral RNAi activity is high in cells lines from two bats species (P.alecto and T. brasiliensis) and that this pathway might play an important role in bat viral tolerance. In this study, the authors investigate the antiviral role of Dicer in another bat species, Myotis myotis. First they express M. myotis Dicer (mmDicer) or human Dicer (hDicer) in a human epithelial kidney (HEK) 293T cell line knockout for Dicer (NoDice cells) and show that, in a human cell line, expression of mmDicer or hDicer doesn't restrict infections with either Sindbis virus (SINV) or vesicular stomatitis virus (VSV). The authors then tested the role of endogenous bat Dicer in M. myotis nasal epithelial cells and found that mmDicer has a proviral activity since its knockdown reduced the replication of SINV and Semliki Forest virus (SFV), but not of VSV. The authors also show by small RNA deep sequencing analysis that there was only a modest RNAi signature in both HEK293T and M. myotis infected with SINV suggesting that mmDicer does not have increased RNAi activity compared to human cells. Interestingly, the authors then found that in M. myotis cells infected with SINV, SFV but not VSV, mmDicer accumulates into cytoplasmic foci, which also contain double-stranded RNA (dsRNA) derived from viral replication. Finally, the authors showed that this relocalisation of mmDicer into foci was dependent on host factors from M. myotis cells as there was no change in localisation in SINV-infected HEK 293T NoDice cells complemented with mmDicer.
Major comments
Minor comments
The findings from this study are interesting as they provide further insights into the role of RNAi towards virus infections. Notably, it highlights a putative proviral role of Dicer in M. myotis bat cells in contrast to the antiviral role in mammals (including other bat species) as well as in plants and invertebrates. Another exciting finding of this study is the observation that mmDicer accumulates in cytoplasmic foci upon viral infection and that these foci also contain viral dsRNA replication intermediates. These accumulation of Dicer into foci only appear in bat cells infected with viruses producing large amounts of dsRNA such as SFV and SINV but not with VSV infection where no dsRNA was detected.
While these findings are novel and interesting, this study, as it stands, is rather descriptive and doesn't provide mechanistic insights into the proviral activity of mmDicer and its localisation into cytoplasmic foci upon infections. The importance of the authors' findings would greatly improve if there were some experiments addressing whether this localisation of mmDicer into foci is responsible or at least correlate with its proviral activity/its lack of antiviral activity. Comparative studies between M. myotis cells in which Dicer is proviral and/or P.alecto and T. brasiliensis cells where RNAi was previously shown to be antiviral would likely provide key mechanistic insights.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Manuscript number: RC-2025-02946
Corresponding author(s): Margaret, Frame
Roza, Masalmeh
[Please use this template only if the submitted manuscript should be considered by the affiliate journal as a full revision in response to the points raised by the reviewers.
If you wish to submit a preliminary revision with a revision plan, please use our "Revision Plan" template. It is important to use the appropriate template to clearly inform the editors of your intentions.]
This section is optional. Insert here any general statements you wish to make about the goal of the study or about the reviews.
We thank the reviewers for recognizing the significance of our work and for their constructive feedback and suggestions, most of which we have implemented in our revised manuscript.
This section is mandatory. *Please insert a point-by-point reply describing the revisions that were already carried out and included in the transferred manuscript. *
Reviewer #1
Evidence, reproducibility and clarity
Review of Masalmeh et al. Title: "FAK modulates glioblastoma stem cell energetics..."
Previous studies have implicated FAK and the related tyrosine kinase PYK2 in glioblastoma growth, cell migration, and invasion. Herein, using a murine stem cell model of glioblastoma, the authors used CRISPR to inactivate FAK, FAK-null cells selected and cloned, and lentiviral re-expression of murine FAK in the FAK-null cells (termed FAK Rx) was accomplished. FAK-/- cells were shown to possess epithelial characteristics whereas FAK Rx cells expressed mesenchymal markers and increased cell migration/invasion in vitro. Comparisons between FAK-/- and FAK Rx cells showed that FAK re-expressed increased mitochondrial respiration and amino acid uptake. This was associated with FAK Rx cells exhibiting filamentous mitochondrial morphology (potentially an OXPHOS phenotype) and decreased levels of MTFR1L S235 phosphorylation (implicated in mito morphology fragmentation). Mito and epithelial cell morphology of FAK-/- cells was reversed by treatment with Rho-kinase inhibitors that also increased mito metabolism and cell viability. Last, FAK-dependent glioblastoma tumor growth was shown by comparisons of FAK-/- and FAK Rx implantation studies.
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
Some questions that would enhance potential impact. 1. Cell generation. Please describe the analysis of FAK-/- clones in more detail. The "low viability" phenotype needs further explanation with regard to clonal expansion and growth characteristics?
Response:
Figure 1F: need further support of MET change upon FAK KO and EMT reversion.
Response: We have added a heatmap (Figure S1E) illustrating the changes in protein expression of core-enriched EMT/MET genes products (by proteomics) after FAK gene deletion (EMT genes as defined in Howe et al., 2018) ; this strengthens the conclusion that the MET reversion morphological phenotype is accompanied by recognised MET protein changes.
Fig. 2: Need further support if FAK effects impact glycolysis or oxidative phosphorylation in particular as implicated by the stem cell model.
Response: We show that FAK impacts both glycolysis (Figure 2A, 2E, and 2F) and mitochondrial oxidative phosphorylation on the basis of the oxygen consumption rate (OCR) (Figure 2B, and 2D), showing both are contributing pathways to FAK-dependent energy production. We have clarified this in the text.
Is there a combinatorial potential between FAKi and chemotherapies used for glioblastoma. Need to build upon past studies.
Response: Yes, previous studies suggest that inhibiting FAK can sensitize GBM cells to chemotherapy (Golubovskaya et al., 2012; Ortiz-Rivera et al., 2023). We have included a paragraph in the discussion section to make sure this is clearer. Although it is not the subject of this study, we appreciate it is useful context.
The notation of changes in glucose transporter expression should be followed up with regard to the potential that FAK-expressing cells may have different uptake of carbon sources and other amino acids. Altered uptake could be one potential explanation for increase glycolysis and glutamine flux.
Response: We agree with the reviewer that glucose uptake could be contributing and we include data that 2 glucose transporters are indeed FAK-regulated namely Glucose transporter 1 (GLUT1, encoded by Slc2a1 gene) and Glucose transporter 3 (GLUT 3, encoded by Slc2a3 gene) (shown in Figure S2B and C).
It would be helpful to support the confocal microscopy of mitos with EM.
Response:
We are concerned (and in our experience) that Electron microscopy (EM) may introduce artefacts during sample preparation. In contrast, immunofluorescence sample preparation is less susceptible to artefacts. The SORA system we used is not a conventional point-scanning confocal microscope, but is a super-resolution module based on a spinning disk confocal platform (CSU-W1; Yokogawa) using optical pixel reassignment with confocal detection. This method enhances resolution in all dimensions with resolution in our samples measured at 120nm. This has been instructive in defining a new level of changes in mitochondrial morphology upon FAK gene deletion.
Lack of FAK expression with increased MTFR1 phosphorylation is difficult to interpret.
Response: We do not directly show that this phosphorylation event is causal in our experiments; however, we think it important to document this change since it has been published that phosphorylation of MTFR1 has been causally linked to the mitochondrial morphology we observed in other systems (Tilokani et al., 2022).
Need to have better support between loss of FAK and the increase in Rho signaling. Use of Rho kinase inhibitors is very limited and the context to FAK (and or Pyk2) remains unclear. Past studies have linked integrin adhesion to ECM as a linkage between FAK activation and the transient inhibition of RhoA GTP binding. Is integrin signaling and FAK involved in the cell and metabolism phenotypes in this new model?
Response: To better support the antagonistic effect of FAK on Rho-kinase (ROCK) signalling, we included a new experiment in which the integrin-FAK signalling pathway has been disrupted by treating FAK WT cells with an agent that causes detachment from the substratum, Accutase, and growing the cells in suspension in laminin-free medium. We present ROCK activity data, as judged by phosphorylated MLC2 at serine 19 (pMLC2 S19), relating this to induced FAK phosphorylation at Y397 (a surrogate for FAK activity) that is supressed after integrin disengagement. These measurements have been compared with conditions whereby integrin-FAK signalling is activated by growing the cells on laminin coated surfaces. We observed a time-dependent decrease in pFAK(Y397) levels (normalised to total FAK) in suspended cells compared to those spread on laminin, while pMLC2(S19) levels increased in a reciprocal manner over time in detached cells relative to spread cells (S4A and B). There is therefore an inverse relationship between integrin-FAK signalling and ROCK-MLC2 activity, consistent with findings from FAK gene deletion experiments. In the former case, we do not rely on gene deletion cell clones.
Significance
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
__Response: __
Deleting the gene encoding FAK in mouse embryonic fibroblasts leads to elevated Pyk2 expression (Sieg, 2000). However, in the GBM stem cell model we used here, Pyk2 was not expressed (determined by both transcriptomics and proteomics). We have included Figure S1E to show that PYK2 expression was undetectable in FAK -/- and FAK Rx cells at the RNA level (Figure S1F). We conclude that there is no compensatory increase in Pyk2 upon FAK loss in these cells. In the transformed neural stem cell model of GBM, we do not consistently or robustly detect nuclear FAK.
Review #2
Masalmeh and colleagues employ a neural stem/progenitor cell-based glioma model (NPE cells) to investigate the role of Focal Adhesion Kinase (FAK) in GBM, with a focus on potential links between the regulation of morphological/adhesive and metabolic GBM cell properties. For this, the authors employ wt cells alongside newly generated FAK-KO and -reexpressing cells, as well as pharmacological interventions to probe the relevance of specific signaling pathways. The authors´ main claims are that FAK crucially modulates glioma cell morphology, cell-cell and cell-substrate interactions and motility, as well as their metabolism, and that these effects translate to changes to relevant in vivo properties such as invasion and tumor growth.
My main issues are with the model chosen by the authors.
As per the methods section, generation of FAK-KO and -"Rx" NPE cells entailed protracted selection/expansion processes, which may have resulted in inadvertent selection for cellular/molecular properties unrelated to the desired one (loss or gain of FAK expression) and which may have had cascading effects on NPE cells. The authors nonetheless repeatedly claim the parameters they quantify, such as mitochondrial or cytoskeletal properties or metabolic features, to have directly resulted from FAK loss or reintroduction. Examples of such causal inferences are to be found in lines 123, 134/135, 165, 181. Such causal claims are, in my view, unsupported.
Acute perturbation of FAK expression/activity, genetically or pharmacologically, followed by a rapid assessment of the processes under investigation, would be needed to begin to assess causality, even if acute genetic perturbations may be technically challenging as sufficient gene expression reduction or restoration to physiologically relevant levels may be hard to achieve.
Response:
We would like to first comment on the model we used here, which we think will clarify the validity of our approach. The model is a transformed stem cell model of GBM that was published in (Gangoso et al., Cell, 2021) and is now used regularly in the GBM field. As mentioned in the response to Reviewer 1, we have added text (page 4 and 5 in the revised manuscript) and a new supplementary figure (Figure S1D) clarifying that the morphological changes we observed were consistent across multiple FAK -/- clones, showing this was not due to any inter-clonal variability. We also added images showing that the morphological changes were apparent at 48 h after nucleofecting FAK -/- cells with the FAK‑expressing vector specifically (not the empty vector), prior to starting G418 selection to enrich for FAK‑expressing cells (Figure S1C), addressing the worry that clonal variation and selection was the cause of the FAK-dependent phenotypes we observed. We believe that our model provides a type of well controlled, clean genetic cancer cell system of a type that is commonly used in cancer cell biology, allowing us to attribute phenotypes to individual proteins.
We have also carried out a more acute treatment by using the FAK inhibitor VS4718 to perturb FAK kinase activity and assessed the effects on glycolysis and glutamine oxidation after 48h treatment (Figure S2D, E and F). We found that treating the transformed neural stem cells (parental population) with FAK inhibitor (300nM VS4718) decreases glucose incorporation into glycolysis intermediates and glutamine incorporation into TCA cycle intermediates, consistent with a role for FAK's kinase activity in maintaining glycolysis and glutamine oxidation.
The employed pharmacological modulation of ROCK activity is the only approach that, given the presumably acute nature of the treatment, may have allowed the authors to probe the proposed functional links. The methods section of the manuscript does not however comprise details as to the duration of these treatments, which leaves open the possibility of long-term treatment having been carried out (data shown in Figure 5B refers to 72hr treatment).
__Response: __
We have added the duration of the treatment to the Methods section and Figure Legends, to clarify that cells were treated with ROCK inhibitors for 24h, before assessing the effects on mictochondria (Figure 4C, D, S4C and D) and glutamine oxidation (Figure 5A, and S5). For metabolic activity by AlamarBlue assay, cells were treated with ROCK inhibitors for 72h (Figure 5B).
Even in the case of ROCK inhibitor experiments, it is however unclear if and how the effects on cell morphology and adhesion, mitochondrial organization and metabolic activity may be connected to each other and, if at all, to FAK expression.
Given the above uncertainties due to the nature of the model and experimental approaches, it is hard to assess the reliability and thus the relevance of the findings.
Response:
FAK suppresses ROCK activity (as judged by pMLC2 S19, Figure 4A and B). Treating FAK -/- cells with two different ROCK inhibitors restored mesenchymal-like cell morphology, mitochondrial morphology and glutamine oxidation. As mentioned above, to strengthen our evidence for the antagonistic role of FAK in ROCK-MLC2 signalling, we have now introduced an experiment whereby integrin-FAK signalling was disrupted through treatment with a detachment agent (Accutase), and subsequently maintaining the cells in suspension in laminin-free medium. We assessed pMLC2 S19 levels (a measure of ROCK activity) relating this to FAK phosphorylation that is supressed after integrin disengagement. These results were evaluated relative to spread wild type cells growing on laminin where Integrin-FAK signalling was active (Figure S4A and B). We observed an inverse relationship between Integrin-FAK signalling and ROCK-MLC2 activity in keeping with our conclusions (Figure 4A and B).
Experimental support for the ability of cell-substrate interaction modulation to concomitantly impact cellular metabolism and motility/invasion would be significant both in terms of advancing our understanding of glioma cell biology and of its translational potential, but the evidence being provided is at best compatible with the proposed model.
Response: We carried out a new experiment to support the ability of cell-substrate interaction modulation to impact metabolism; specifically, we inhibited cell-substrate interactions by plating the cells on Poly-2-hydroxyethyl methacrylate (Poly 2-HEMA)-coated dishes. This suppressed FAK phosphorylation at Y397, as expected, with concomitant reduction in glutamine utilisation in the TCA cycle (Figure S3A, B and C).
My background/expertise is in developmental and adult neurogenesis, in vivo modelling of gliomagenesis and cell fate control/reprogramming, with a focus on molecular mechanisms of differentiation and quantitative aspects of lineage dynamics; molecular details of the control of cellular metabolism, cell-cell adhesion and cytoskeletal dynamics are not core expertise of mine.
We appreciate this reviewer's expertise are not necessarily in the cancer cell biology and genetic intervention aspects of our study. We hope that the explanations we have provided satisfy the reviewer that our conclusions are valid.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Masalmeh and colleagues employ a neural stem/progenitor cell-based glioma model (NPE cells) to investigate the role of Focal Adhesion Kinase (FAK) in GBM, with a focus on potential links between the regulation of morphological/adhesive and metabolic GBM cell properties. For this, the authors employ wt cells alongside newly generated FAK-KO and -reexpressing cells, as well as pharmacological interventions to probe the relevance of specific signaling pathways. The authors´ main claims are that FAK crucially modulates glioma cell morphology, cell-cell and cell-substrate interactions and motility, as well as their metabolism, and that these effects translate to changes to relevant in vivo properties such as invasion and tumor growth. My main issues are with the model chosen by the authors.
As per the methods section, generation of FAK-KO and -"Rx" NPE cells entailed protracted selection/expansion processes, which may have resulted in inadvertent selection for cellular/molecular properties unrelated to the desired one (loss or gain of FAK expression) and which may have had cascading effects on NPE cells. The authors nonetheless repeatedly claim the parameters they quantify, such as mitochondrial or cytoskeletal properties or metabolic features, to have directly resulted from FAK loss or reintroduction. Examples of such causal inferences are to be found in lines 123, 134/135, 165, 181. Such causal claims are, in my view, unsupported. Acute perturbation of FAK expression/activity, genetically or pharmacologically, followed by a rapid assessment of the processes under investigation, would be needed to begin to assess causality, even if acute genetic perturbations may be technically challenging as sufficient gene expression reduction or restoration to physiologically relevant levels may be hard to achieve.
The employed pharmacological modulation of ROCK activity is the only approach that, given the presumably acute nature of the treatment, may have allowed the authors to probe the proposed functional links. The methods section of the manuscript does not however comprise details as to the duration of these treatments, which leaves open the possibility of long-term treatment having been carried out (data shown in Figure 5B refers to 72hr treatment). Even in the case of ROCK inhibitor experiments, it is however unclear if and how the effects on cell morphology and adhesion, mitochondrial organization and metabolic activity may be connected to each other and, if at all, to FAK expression.
Given the above uncertainties due to the nature of the model and experimental approaches, it is hard to assess the reliability and thus the relevance of the findings.
Experimental support for the ability of cell-substrate interaction modulation to concomitantly impact cellular metabolism and motility/invasion would be significant both in terms of advancing our understanding of glioma cell biology and of its translational potential, but the evidence being provided is at best compatible with the proposed model.
My background/expertise is in developmental and adult neurogenesis, in vivo modelling of gliomagenesis and cell fate control/reprogramming, with a focus on molecular mechanisms of differentiation and quantitative aspects of lineage dynamics; molecular details of the control of cellular metabolism, cell-cell adhesion and cytoskeletal dynamics are not core expertise of mine.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Review of Masalmeh et al.
Title: "FAK modulates glioblastoma stem cell energetics..."
Previous studies have implicated FAK and the related tyrosine kinase PYK2 in glioblastoma growth, cell migration, and invasion. Herein, using a murine stem cell model of glioblastoma, the authors used CRISPR to inactivate FAK, FAK-null cells selected and cloned, and lentiviral re-expression of murine FAK in the FAK-null cells (termed FAK Rx) was accomplished. FAK-/- cells were shown to possess epithelial characteristics whereas FAK Rx cells expressed mesenchymal markers and increased cell migration/invasion in vitro. Comparisons between FAK-/- and FAK Rx cells showed that FAK re-expressed increased mitochondrial respiration and amino acid uptake. This was associated with FAK Rx cells exhibiting filamentous mitochondrial morphology (potentially an OXPHOS phenotype) and decreased levels of MTFR1L S235 phosphorylation (implicated in mito morphology fragmentation). Mito and epithelial cell morphology of FAK-/- cells was reversed by treatment with Rho-kinase inhibitors that also increased mito metabolism and cell viability. Last, FAK-dependent glioblastoma tumor growth was shown by comparisons of FAK-/- and FAK Rx implantation studies.
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
Some questions that would enhance potential impact.
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
RESPONSE TO REVIEWERS
We thank the reviewers for their thoughtful and constructive feedback, which has been instrumental in improving the overall quality of our manuscript.
In response, we have undertaken a substantial revision that includes new experimental data, refined analyses, and clearer presentation of our findings. Specifically, we have addressed concerns about RNAi efficiency and protein-level validation, expanded our genetic models to include loss-of-function contexts, and clarified the interpretation of mitochondrial morphology using both confocal and electron microscopy. We also incorporated new data on Cyclin E regulation and mitochondrial membrane potential to strengthen the mechanistic link between dPGC1 depletion and Yki-driven tumorigenesis. These revisions not only address the specific points raised by the reviewers but also enhance the coherence and impact of the study. We are confident that the revised manuscript presents a more robust and compelling case for the role of dPGC1 as a context-dependent tumor suppressor and that it will be of broad interest to the fields of developmental biology, cancer metabolism, and mitochondrial dynamics.
Reviewer #1 (Evidence, reproducibility and clarity (Required)): Sew et al. examine the master regulator of mitochondrial biogenesis, dPGC1, in the context of Drosophila wing and larval development. They primarily use confocal imaging to probe the interplay between dPGC1 and an overactive Hippo pathway, driven by overexpression of the main effector protein, Yki. In their study, they find that tumors, driven by overactivity of Yki grow larger when dPGC1 is downregulated, implicating the mitochondrial biogenesis pathway in tumor suppression. Furthermore, in the context of Yki overexpression, they find that levels of Mfn or Opa1 modulate tumor size. Lastly, they show a role of cyclin E in controlling the size of tumors formed by Yki OE + dPGC1 RNAi. The potential role of dPGC1 as a tumor suppressor is interesting because it highlights an emerging recognition of mitochondria in the aetiology of cancer. However, before publication, much of the data in this manuscript should be strengthened by a refinement in the methods/analysis and an increase in orthogonal approaches.
We addressed concerns regarding RNAi efficiency and wing development by incorporating data from a dPGC1 mutant allele and using a ubiquitous driver for qPCR validation of transgene efficiency. We clarified the rationale for EM use. The manuscript now avoids overinterpretation of mitochondrial morphology and focuses on fusion-specific regulators. We also revised the narrative arc to maintain coherence and added loss-of-function models to support our conclusions.
Below, we address each of the reviewer’s points in detail.
Major comments:
The authors indicate that for example, in lines127-28, that neither downregulating or overexpressing dPGC1 affects wing size. However, the quantification in Fig. 1C shows a significant decrease in wing size following RNAi treatment. This decrease is modest, but it is nevertheless significant. It is worth pointing out, too, that the efficiency of the RNAi in Fig. S1C suggests that the conclusions drawn are premature. While a roughly 55% drop in mRNA levels may be statistically significant, it is unclear whether this drop in transcripts corresponds to a commensurate depletion of protein. Moreover, it is unclear, in this context, how much dPGC1 may indeed be necessary to drive a relatively normal program of mitochondrial biogenesis in wing development. To obtain a clear result, it is necessary to show significant depletion of the dPGC1 protein. (Ultimately, if it is the case that dPGC1 is unnecessary for wing development and function, a more coherent line of inquiry would be to find out the reason for this rather than to pivot the story to studying tumorigenesis in larva.)
We agree that the interpretation of the RNAi efficiency data requires clarification.
The qPCR analysis shown in former Fig. S1C was performed using wing discs from flies expressing UAS-dPGC1-RNAi under the control of the MS1096-Gal4 driver. However, as shown in current Fig. 1C, MS1096-Gal4 is not expressed uniformly across the wing disc. Some regions remain RFP-negative, indicating that the RNAi construct is not active in all cells. As a result, the measured mRNA levels likely underestimate the true knockdown efficiency. This is because the qPCR includes mRNA from both RNAi-expressing and non-expressing cells, diluting the apparent reduction in transcript levels.
To address this limitation and more accurately assess RNAi efficiency, we repeated the qPCR analysis using a ubiquitous driver (actin-Gal4) to ensure uniform expression of the RNAi construct. Under these conditions, we observed a more substantial knockdown, with dPGC1 mRNA levels reduced to approximately 25% of control levels (this is shown in current Fig S2). This result indicates that the RNAi line is more effective than initially suggested by the MS1096-Gal4-based analysis.
To complement our RNAi-based analysis, we additionally used a mutant strain carrying a characterized allele of dPGC1 (dPGC11, also known as dPGC1KG08646; see FlyBase: https://flybase.org/reports/FBal0148128). This genetically distinct approach allowed us to validate and strengthen our findings regarding dPGC1 function. Flies homozygous for this allele exhibited a modest but statistically significant reduction in both wing disc and adult wing size. These results support the conclusion that dPGC1 is required for normal wing growth and development. The new data are now included in Figure 1 and referenced in the main text (lines 144-153).
Additionally, as suggested by the reviewer, we have revised the relevant section to maintain a coherent line of inquiry. The updated text can be found in lines 163–172.
In Figure 3H-K, it is not clear why the authors used electron microscopy to evaluate mitochondrial morphology. The very good confocal images in Figure 3C-G show a clear change in mitochondrial morphology following the knockdown of Mfn, Opa1, and Miro. While it is clear from the electron micrographs in Figure H that the mitochondria are enlarged, it is not obvious that this increase in length is a result of increased mitochondrial fusion. Indeed, if the mean form factor were used to quantify the shape, it is likely that in both conditions, the value would be close to 1, indicating more of a round object, and it not obvious whether there would be a difference between the Yki OE versus the YkI OE + dPGC1 RNAi. Therefore, from this data alone, it cannot be concluded that the YkI OE + dPGC1 RNAi condition leads to mitochondrial hyperfusion.
Our rationale for including electron microscopy (EM) was to overcome specific limitations in imaging mitochondrial morphology within the main epithelium of the wing disc, where Yki-driven tumors arise. These tumors were generated using ap-Gal4, which drives expression specifically in the main epithelium and is not active in the peripodial membrane. This is an important distinction, as the peripodial membrane—used in Figures 3C–G—has a squamous architecture and larger cytoplasmic volume, making it ideal for high-resolution confocal imaging and for assessing the effects of manipulating dMfn, Opa1, and miro. However, because ap-Gal4 is not expressed in the peripodial membrane, this tissue could not be used to analyze mitochondrial morphology in the actual tumorous context.
To directly evaluate mitochondria in the main epithelium, we employed EM, which provides the resolution necessary to visualize ultrastructural changes that are not easily captured by confocal microscopy in this densely packed tissue. While EM does not directly measure fusion events, it allowed us to detect changes in mitochondrial size and shape that support our broader findings.
We acknowledge that mitochondrial enlargement alone does not definitively demonstrate hyperfusion. However, the EM data were interpreted alongside additional evidence: the upregulation of mitochondrial fusion genes (dMfn and Opa1) in Yki + dPGC1-RNAi tumors, and functional data showing that overexpression of these genes promotes fusion in the peripodial membrane. Together, these findings suggest that dPGC1 depletion enhances mitochondrial fusion in Yki-driven tumors.
To further clarify this point, we also imaged mitochondria in the main epithelium using confocal microscopy. However, the resolution was considerably lower than that achieved with EM, limiting our ability to assess fine mitochondrial structures. We have prepared a representative figure for the reviewer (below), showing representative confocal images of wing discs from three genotypes: (A) ap-Gal4, UAS-GFP (control), (B) ap-Gal4, UAS-Yki, and (C) ap-Gal4, UAS-Yki, UAS-dPGC1-RNAi. We used anti-ATP-synthase (Abcam, ab14748, dilution 1:200), to label the mitochondria for this Figure. Despite the lower resolution, mitochondria in the Yki + dPGC1-RNAi tumors appear elongated (yellow arrows) compared to those in the other conditions, consistent with the changes observed by EM. We believe this example illustrates the limitations of confocal imaging in this tissue and reinforces the need for EM to accurately assess mitochondrial morphology in the tumorous epithelium.
While our EM analyses reveal mitochondrial enlargement in wing discs co-expressing Yki and PGC1-RNAi, we acknowledge that these structural features alone do not conclusively demonstrate mitochondrial hyperfusion. To address this, we have revised the manuscript to avoid overinterpreting the EM data and instead emphasize the functional relevance of mitochondrial fusion regulators such as dMfn and Opa1 in promoting tumor growth.
Taken together, the EM analysis provides structural validation in the tumorous epithelium (Fig 4), while the confocal imaging and functional manipulation of fusion genes in the peripodial membrane offer mechanistic insight (Fig 3). This integrated approach strengthens the conclusion that PGC1 depletion in a Yki-overexpressing context promotes changes in mitochondrial morphology and contributes to tumorigenesis, independent of whether these changes reflect hyperfusion.
Figure 4. refers to changes in mitochondrial fusion and fission in tumor formation; however, the authors do not attempt to alter mitochondrial fission factors, so it is not accurate to mention a role of mitochondrial fission, in this context.
As we did not directly manipulate fission-related factors in our experiments, we agree that it would be inappropriate to draw conclusions about the role of mitochondrial fission in this context. Our revised figure (current Fig 5) and accompanying text now focus exclusively on the effects of mitochondrial fusion and the genes directly involved in regulating this process.
It must be noted, too, that the authors have not demonstrated that their genetic interventions have actually affected mitochondrial morphology in these experiments. As noted in the previous figure, the Yki OE + dPGC1 RNAi condition showed enlarged mitochondria, but not necessarily hyperfused organelles. Therefore, the downregulation of Mfn or Opa1 in this set of experiments may not necessarily have altered mitochondrial morphology. Perhaps suppression of Mfn or Opa1 would normalize the areas of these evidently swollen mitochondria, but this is unclear without images. Furthermore, it should be appreciated that both Opa1 and Mfn exhibit pleiotropic attributes - e.g., Opa1 not only regulates IMM fusion, but it also modulates the shape and tightness of cristae membranes, specialized sites of oxidative phosphorylation as well as sequestration of cytochrome c, the release of which influences apoptosis (Frezza et al., 2006). At least in mammalian cells, Mfn2 is thought to regulate contacts between mitochondria and endoplasmic reticulum (Naon et al., 2023), which may serve other functions than OMM fusion, such as stabilization of the MAM.
To directly address this point, we performed EM to assess mitochondrial ultrastructure in Yki + dPGC1-RNAi wing disc tumors, with and without dMfn1 downregulation, the most upregulated mitochondrial fusion gene in this tumor context. In Yki + dPGC1-RNAi tumors, mitochondria appeared more elongated, consistent with increased fusion. Upon dMfn1 depletion, we observed a dramatic shift in mitochondrial morphology: mitochondria became larger and more rounded, with disrupted cristae and onion-like structures, indicative of compromised mitochondrial integrity and function (see current Fig. 4).
As the reviewer rightly notes, these morphological changes are consistent with the pleiotropic roles of Mfn and Opa1, which extend beyond outer and inner membrane fusion to include regulation of cristae architecture and ER-mitochondria contacts (Frezza et al., 2006; Naon et al., 2023). We now discuss these broader roles in the revised manuscript (lines 493–497). Taken together, our EM and confocal analyses, combined with targeted genetic manipulations, provide evidence that mitochondrial morphology is indeed altered in response to dPGC1 depletion and fusion gene deregulation in the wing disc.
Figure 5 highlights a connection between dysregulation of mitochondria and Cyclin E, which allows cells to prematurely enter S phase. The data presented here do not offer clarity on whether the enlargement of the tumors results from increase cellular proliferation and/or cell size. The role of the cell cycle adds a layer of complexity to these results, because it is thought that mitochondria undergo fragmentation during the cell cycle to promote an even distribution of the organelle population after mitosis (Taguchi et al., 2007); however, in this manuscript, the authors contend that the downregulation dPGC1 is promoting mitochondrial hyperfusion. It is unclear how and whether cellular division and proliferation would proceed at an accelerated rate in a situation with mitochondrial hyperfusion.
To address this point, we started by analyzing whether Yki + dPGC1-RNAi tumors exhibit increased proliferation compared to tumors expressing Yki alone. We quantified mitotic activity using the phospho-Histone H3 (PH3) marker of mitotic cells and observed a significant increase in PH3-positive cells in the Yki + dPGC1-RNAi condition. These results indicate an elevated proliferation rate in these tumors and are now presented in Fig 2O–Q. In the text, can be found in lines 221-228.
We agree with the reviewer that our findings challenge the conventional view that mitochondrial fragmentation is a prerequisite for mitosis, as we observe increased expression of gene promoting mitochondrial fusion in the context of dPGC1 downregulation alongside signs of accelerated cell cycle entry. It is important to note that we also show that the levels of the oncogene Cyclin E, a key driver of cell cycle progression and S-phase entry, were elevated in Yki + dPGC1-RNAi tumors compared to those expressing Yki alone, suggesting that the increased proliferation observed is at least in part driven by enhanced cycle activity. To further probe Cyclin E’s role, we used the CycE-05306 heterozygous mutant allele, which reduces Cyclin E levels by ~50% without affecting normal development. Notably, this partial reduction strongly suppressed tumor growth in the Yki + dPGC1-RNAi background (Fig 6), underscoring Cyclin E’s functional importance in supporting oncogenic growth in this context.
These findings support the notion that defects in the expression of mitochondrial genes involved in mitochondrial morphology induced by dPGC1 depletion do not impair but rather coincide with accelerated cell division.
Minor comments:
Lines 69-72 contrast the roles of PGC1α and β. It is not clear whether the comparison is of their respective roles in cancer or in normal physiology. In either case, it is important to note that PGC1β has been shown to drive mitochondrial fusion as well as biogenesis through its control of MFN2, among other factors (Liesa et al., 2008).
In response, we have clarified the comparison between PGC1α and PGC1β in the introduction to specify that it refers to their roles in cancer. Additionally, we now acknowledge that PGC1β has been shown to promote mitochondrial biogenesis and fusion, notably through the regulation of MFN2, as demonstrated by Liesa et al. (2008). This reference has been added to provide a more balanced and accurate representation of PGC1β’s functions. In the text it can be found in lines 77-81.
Although this study focuses on PGC1, the authors do not seem to site the original literature from the Spiegelman lab.
In response to the reviewer’s comment, we have added a new section in the introduction that cites key foundational studies from the Spiegelman lab. This addition can be found in the introduction in lines 68-73.
There are 10-20 grammatical errors throughout the text.
We apologize for this. We have carefully revised the text, and we are very confident those errors have been corrected.
**Referee Cross-commenting**
There is agreement among the referees that the potential role of PGC1 as a tumor suppressor is interesting and significant. However, various aspects of this work require attention prior to publication. For example, there needs to be a complete knock down of PGC1 to come to any conclusion as to its role in wing development. The methods for analyzing mitochondrial morphology need to be clarified and be consistent with standards in the field of mitochondrial dynamics. Also, the authors need to quantify their Western blots to obtain accurate assessments of protein levels. Generally, the study relies too heavily on overexpression experiments; understanding the potential role of mitochondria in regulating the Hippo pathway should include various knockdown and/or knockout models.
Reviewer #1 (Significance (Required)):
Overall, the authors show an interesting dampening effect of dPGC1 on growth of Yki-driven tumors. This data could be relevant for elucidating how dysregulation of the Hippo signalling pathway can underlie tumorigenesis.
The narrative arc of the study, however, appears to lack a focused line of inquiry. Figure 1 highlights an attempt to modulate Drosophila wing size and/or structure by downregulating dPGC1, but to no effect. Although examination of the efficiency of the RNAi revealed that the transcripts were still present in significant quantities; so, the conclusion that dPGC1 is dispensable for wing formation is premature. To have clarity on this point, it would be necessary to completely knockdown the gene, preferably by showing a total loss of protein. This should be feasible for the authors, since they showed Western blotting in Figure 5A. In any event, it seems that this negative data led the authors to study the Hippo pathway in the larval stage. This transition from Figure 1 to 2 seemed somewhat arbitrary and leads to a rather disjointed sense of the main line of inquiry around dPGC1.
It is important to note, too, that the authors highlight a role of mitochondrial dynamics in the pathway of Yki-driven tumor formation; however, they only directly evaluate mitochondrial dynamics in this context in a single assay, namely, Figure 3H-K, and this quantification is likely inaccurate because the mitochondria in the Yki OE + dPGC1 RNAi condition seem to be substantially enlarged, circular structures. It is critical to keep in mind that mitochondrial enlargement does not necessarily stem from hyperfusion. It could come from a decrease in the activity of Drp1 or result from an imbalance between mitochondrial biogenesis and mitophagy.
As noted in our responses above, we have addressed these concerns by clarifying the limitations of our mitochondrial morphology analysis. Additionally, we have expanded the discussion (lines 498-504) to explicitly acknowledge that mitochondrial enlargement does not necessarily indicate hyperfusion. In that paragraph, we consider alternative explanations such as reduced fission or imbalances in mitochondrial biogenesis and mitophagy, and we outline the need for future studies using dynamic assays and additional markers to more precisely dissect mitochondrial remodeling in Yki-driven tumors.
A marked limitation of this study is the overuse of rather artificial manipulations of transcriptional regulatory pathways. The study would benefit a lot from investigation of the loss of function of components of the Hippo pathway rather than just OE of Yki.
We performed additional experiments using Warts (Wts) mutant clones to assess the role of dPGC1 in a loss-of-function context within the Hippo pathway. While our initial analyses were based on Yki overexpression, which allowed us to robustly probe the interaction between Yki and dPGC1, we agree that this approach may not fully reflect physiological conditions. By generating Wts mutant clones, which endogenously activate Yki through loss of upstream inhibition, we were able to evaluate the impact of dPGC1 depletion in a more physiologically relevant setting. These new results confirm and extend our previous findings, showing that dPGC1 limits tissue overgrowth even when Yki is activated through loss of Wts, thereby strengthening the biological relevance of our conclusions.
These results are presented in Fig 2F-I. In the text, those results are presented in lines 181-189.
My expertise is in mitochondrial biology, with specialization in super-resolution imaging, mitochondrial dynamics and membrane architecture. I have also worked in the interface between mitochondrial physiology and cancer. With this perspective, I think that the authors uncover a potentially interesting role of PGC1 as a tumor suppressor.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
Summary In this manuscript the authors the investigate the role of the mitochondrial regulatory transcription factor dPGC1 in tissue growth and oncogenic transformation. They show that dPGC1 limits hyperplasia mediated by overexpression of Yki in the Drosophila wing disc, while having no effect on normal growth. dPGC1 depletion in discs overexpressing Yki results neoplastic overgrowth and hyperfused mitochondria, which was dependent on the increased expression of genes involved in promoting mitochondrial fusion. Additionally, the authors show that dPGC1 limits CycE levels post-transcriptionally in Yki tumors.
In the revised version of our manuscript, we have clarified the relationship between our findings and prior work by Nagaraj et al., including new experiments that demonstrate the specificity of dPGC1’s role in Yki-driven growth. Specifically, we show that dPGC1 depletion does not enhance tissue overgrowth in EGFR or InR contexts, nor does it affect Yki expression or activity. Furthermore, we tested dPGC1 overexpression in Yki-overexpressing tissues and observed no significant changes in growth or mitochondrial fusion gene expression. Additional controls confirmed that Cyclin E upregulation is specific to the Yki + dPGC1 depletion condition, reinforcing the context-dependent nature of our findings.
Each of the reviewer’s comments is addressed below.
Major comments 1) The authors mention several times in passing in the results a manuscript from the Banerjee lab (Nagaraj et al 2012), which shows that many of the genes the authors of the present manuscript show are upregulated upon Yki overexpression + dPGC1-RNAi compared with Yki overexpression alone are in fact upregulated upon Yki overexpression alone compared with control (dMfn/marf, opa1, miro - while interestingly dPGC1 itself is not affected). Nagaraj et al further show that Yki-overexpressing discs have longer mitochondria suggesting increased fusion even in the absence of dPGC1 depletion. The findings from Nagaraj et al should be mentioned explicitly in the introduction and the relationship between this manuscript and the present work clearly outlined in the discussion.
In the revised manuscript, we have now explicitly referenced the findings of Nagaraj et al. (2012) in the Introduction (lines 106-118), Results (lines 355-360) and Discussion (lines 466-468) sections.
In the revised Introduction, we summarize their key observations that Yki overexpression alone upregulates mitochondrial fusion genes such as dMfn and Opa1, and leads to mitochondrial elongation, while not affecting dPGC1 expression.
In the revised Results section, we mention that, building on that work, our study demonstrates that dPGC1 depletion further amplifies this effect, leading to enhanced mitochondrial elongation and tumor growth.
In the revised Discussion, we now explicitly reference the findings by Nagaraj et al. (2012), which demonstrated that Yki overexpression promotes mitochondrial fusion and upregulates key fusion genes. We build upon this work by showing that dPGC1 depletion in a Yki-overexpressing background further enhances mitochondrial fusion gene expression and tumor growth. This supports a model in which dPGC1 acts as a safeguard against Yki-induced mitochondrial remodeling and oncogenesis, reinforcing its role as a context-dependent tumor suppressor.
Importantly, we show that this effect is context-dependent and not observed in otherwise normal tissues, highlighting a sensitized mitochondrial response to Yki activation when dPGC1 is lost. These additions help delineate the novel contribution of our study in identifying dPGC1 as a critical modulator of mitochondrial dynamics and tumorigenesis downstream of Yki.
2) Given that Yki overexpression alone induces mitochondrial fusion and that dMfn/marf and opa1 depletion suppresses Yki-induced overgrowth (Nagaraj et al), does dPGC1 overexpression also suppress Yki-induced overgrowth?
If so, is this correlated with reduction in dMfn/marf and opa1 compared with Yki overexpression alone?
In response, we performed additional experiments to assess whether dPGC1 overexpression influences Yki-driven overgrowth. We also analyzed the expression of mitochondrial fusion genes (dMfn and Opa1) in this context. As shown in new Fig. S8, dPGC1 overexpression in Yki-overexpressing wing discs did not significantly affect tissue growth, nor did it alter the mRNA levels of key fusion regulators, dMfn and Opa1. These findings suggest that the transcriptional upregulation of mitochondrial fusion genes observed upon dPGC1 depletion is not a general consequence of altered dPGC1 levels, but rather a specific response that emerges in the context of Yki activation. We now present and discuss these results in the revised manuscript (lines 278-285), highlighting the sensitized nature of mitochondrial remodeling in an oncogenic environment driven by Yki signaling.
3) One important question raised by this study is: how specific is the effect of dPGC1 depletion to Yki-driven overgrowth? As Yki-driven overgrowth already have increased mitochondrial length, it is possible that Yki-expressing cells are already sensitised to the effects of dPGC1 depletion. Interestingly, Nagaraj et al show that mitochondrial morphology is not affected upon EGFR activation (hyperplasia) or upon scrib and avl depletion (neoplasia). The authors should therefore test if dPGC1 depletion can potentiate the growth of other hyperplasia drivers such as activated EGFR and InR in the wing disc.
We tested whether the growth-suppressive effect of dPGC1 depletion was specific to Yki-driven overgrowth or could also potentiate tissue growth in other oncogenic contexts. Specifically, we downregulated dPGC1 in wing discs overexpressing either EGFR or InR. In both cases, we did not observe any enhancement of tissue overgrowth upon dPGC1 depletion, in contrast to what we observed in Yki-overexpressing discs. These results suggest that the sensitivity to dPGC1 depletion is specific to Yki-driven overgrowth and is not a general feature of hyperplastic growth induced by other oncogenes.
These results are shown in Fig S4 and in lines 195-202.
4) There are a few simple control experiments the authors should provide to clarify the relationship between Yki and dPGC1: - Are Yki levels affected by dPGC1 depletion?
To address the potential regulation of Yki by dPGC1, we performed quantitative PCR (qPCR) analysis to measure the expression levels of yki and its well-established transcriptional targets—Cyclin E, Diap1, and bantam—in wing discs depleted of dPGC1. As shown in Fig. S3, we did not detect significant changes in the transcript levels of yki or its target genes, suggesting that the enhanced phenotype observed upon dPGC1 depletion is unlikely to be driven by increased Yki expression or activity. These results indicate that dPGC1 does not strongly influence Yki expression or activity. These new results are presented in lines 190-194.
We have conducted this analysis, and the results are now presented in new Fig S7. While the trend is similar to that observed in tumors with both Yki depletion and dPGC1 depletion, the magnitude of change is smaller compared to the context of Yki overexpression. This is described in the text in lines 273-277.
To address this, we examined Cyclin E levels in wing imaginal discs mutant for dPGC1 alone. Our analysis did not reveal any detectable changes in Cyclin E levels under these conditions. These findings suggest that the upregulation of Cyclin E is not a general consequence of dPGC1 loss, but rather a feature specific to the context of Yki overactivation. The corresponding data are now included in Fig S14 of the revised manuscript. In the text, it can be found in lines 442-448.
5) Figure 3C-G: it is not clear how the authors can quantify the length of 3D structures like mitochondria from 2D TEM images (unless they have done volume reconstruction from consecutive sections) and no details are provided in the methods. The quantification of mitochondrial length has to be performed rigorously as it is a key part of the paper.
We agree that TEM provides only 2D profiles of 3D mitochondrial structures, and that this does not allow for precise volumetric reconstruction. In our study, we measured the longest axis of mitochondria visible in thin TEM sections, which is a commonly used 2D proxy for mitochondrial length in the literature (e.g., PMID: 36367943 and PMID: 38637532). To avoid misunderstandings, we have clarified in the Material and Methods section that the reported values represent apparent mitochondrial length in 2D sections, not true 3D length. To enhance the accuracy of these estimates, we measured more than three tissues per genotype, multiple regions per tissue, several cells per region, and various fields of view per cell.
Minor Comments:
1) Line 51: "Mitochondria are highly dynamics organelles." should be "Mitochondria are highly dynamic organelles."
We have corrected that mistake. Thanks!
2) Introduction: the authors should summarise the known physiological functions of PGC1α in order to put their findings in context.
We have added a section in the introduction (lines 66-81) summarizing the known physiological functions of PGC1α
3) lines: 121-3: "...depletion of dPGC1...did not have a major impact on adult wing size and shape (Fig 1B, C)." There is a small but statistically significant difference so the authors should state this in the text.
We have revised the text to acknowledge that dPGC1 depletion leads to a modest but statistically significant reduction in wing size. In addition to the original analysis, we have now included further experiments to strengthen this point. Specifically, we analyzed wings from flies homozygous for the dPGC11 allele (also known as dPGC1KG08646; see FlyBase: https://flybase.org/reports/FBal0148128) and confirmed a small but significant reduction in both wing disc and adult wing size compared to controls (this can be found in Fig. 1 and Fig. S1). These results support the conclusion that, although dPGC1 is dispensable for viability and gross morphology, it contributes to normal wing growth. These new results can be found in lines 144-153.
4) Figure 5A (Cyclin E western blot): the authors should show molecular weight markers. In the revised version of our manuscript, we are including the molecular markers as indicated by the reviewer. These can be found in Fig S12.
Reviewer #2 (Significance (Required)):
The manuscript by Sew et al builds on the previous work by Nagaraj et al to explore the role of mitochondrial function in tumors driven by disruption of the Hippo pathway. In particular, the authors identify dPGC1 as a transcription factor that limits Yki-driven mitochondrial fusion and tissue growth. Interestingly, they further show that Yki/PGC1-depleted tumors are highly sensitive to Cyclin E levels, due to post-transcriptional Cyclin E increase. These results further our knowledge of how Yki drives growth and how mitochondria participate in oncogenic transformation. With appropriate revision as outlined above (for example exploring whether the mechanism proposed is Yki-specific), the manuscript will be of broad interest to developmental and cancer biologists.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
The manuscript presents compelling evidence that dPGC1 acts as a context-dependent tumor suppressor in Drosophila by modulating mitochondrial dynamics and limiting Yorkie (Yki)-induced oncogenic growth. By leveraging the Drosophila wing imaginal disc as a model, the authors investigate how dPGC1 depletion exacerbates Yki-driven tissue overgrowth, mitochondrial hyperfusion, Cyclin E upregulation, and DNA damage, leading to tumorigenesis. The study provides valuable insights into the interplay between mitochondrial dynamics and cancer, with implications for understanding metabolic regulation in oncogenesis. While the findings are significant and well-aligned with the field, certain aspects of the experimental design, data presentation, and mechanistic insights require further attention to enhance clarity, reproducibility, and impact. Below, I outline my major concerns and recommendations.
We addressed concerns about RNAi efficiency and protein-level validation with new qPCR data and mutant analysis. We provided EM and confocal evidence of mitochondrial changes. We clarified non-autonomous effects and quantified Mmp1 and F-actin and added data on miro and Opa1 manipulations. Cyclin E quantification was expanded using multiple Western replicates and a validated mutant allele, and we included new data on mitochondrial membrane potential to assess functional consequences.
Our detailed responses to each point raised by the reviewer are provided below.
Major Points
The qPCR analysis shown in former Fig. S1C was performed using wing discs from flies expressing UAS-dPGC1-RNAi under the control of the MS1096-Gal4 driver. However, as shown in current Fig. 1C, MS1096-Gal4 is not expressed uniformly across the wing disc. Some regions remain RFP-negative, indicating that the RNAi construct is not active in all cells. As a result, the measured mRNA levels likely underestimate the true knockdown efficiency. This is because the qPCR includes mRNA from both RNAi-expressing and non-expressing cells, diluting the apparent reduction in transcript levels.
To address this limitation and more accurately assess RNAi efficiency, we repeated the qPCR analysis using a ubiquitous driver (actin-Gal4) to ensure uniform expression of the RNAi construct. Under these conditions, we observed a more substantial knockdown, with dPGC1 mRNA levels reduced to approximately 25% of control levels (this is shown in current Fig S2). This result indicates that the RNAi line is more effective than initially suggested by the MS1096-Gal4-based analysis.
To complement our RNAi-based analysis, we additionally used a mutant strain carrying a characterized allele of dPGC1 (dPGC11, also known as dPGC1KG08646; see FlyBase: https://flybase.org/reports/FBal0148128). This genetically distinct approach allowed us to validate and strengthen our findings regarding dPGC1 function. Flies homozygous for this allele exhibited a modest but statistically significant reduction in both wing disc and adult wing size. These results support the conclusion that dPGC1 is required for normal wing growth and development. The new data are now included in Figure 1 and referenced in the main text (lines 144-151).
Unfortunately, we cannot perform antibody staining due to the unavailability of antibodies against dPGC1.
How does the wing disc look like when dPGC1 is overepressed together with Yki?
In response, we performed additional experiments to assess whether dPGC1 overexpression influences Yki-driven overgrowth. We also analyzed the expression of mitochondrial fusion genes (dMfn and Opa1) in this context. As shown in new Fig. S8, dPGC1 overexpression in Yki-overexpressing wing discs did not significantly affect tissue growth, nor did it alter the mRNA levels of key fusion regulators, dMfn and Opa1. These findings suggest that the transcriptional upregulation of mitochondrial fusion genes observed upon dPGC1 depletion is not a general consequence of altered dPGC1 levels, but rather a specific response that emerges in the context of Yki activation. We now present and discuss these results in the revised manuscript (lines 278-285), highlighting the sensitized nature of mitochondrial remodeling in an oncogenic environment driven by Yki signaling.
In Fig 2D (but also in Fig. 2C) not only cells in the dorsal but also in the ventral comparmtent seem to overproliferate. Either this is a mis-conception or it is a non-autonomous effect from interfering with Yki and dPGC1 in the vertrnal compartment. In either cases, this has to be clarified.
Ventral cells are not labelled by GFP. Fig 3D shows a tumor in which GFP-negative cells are not present, suggesting that they are not overproliferating but rather being eliminated. This phenomenon is consistent with cell competition, a well-characterized process in which transformed or tumorigenic cells outcompete and eliminate neighboring wild-type cells. We have previously described this behavior in wing disc tumors (PMID: 26853367; DOI: 10.1016/j.cub.2015.12.042), and it likely contributes to the expansion of the tumor mass by removing surrounding normal tissue also in this context.
In Fig. 2F-H quantification of Mmp1 and F-actin is missing. Mmp1 is a JNK target, so the authors could do in addition an anti-phospho JNK antibody staining.
In response, we have performed those quantifications. They are now included in Fig 2M, N.
In Fig. 3: how does the mitochondrial network look like in the wing disc periopodial epithelium using the Gug>Yki+dPGC1 genotype? Is it similar to Gug>dMfn or Gug>miro?
We attempted to perform this analysis; however, Yki overexpression under the control of Gug-GAL4 resulted in larval lethality, likely due to GAL4 activity in essential tissues such as the central nervous system. As a result, we were only able to induce transgene expression for 24 hours before lethality occurred.
At this early point, no detectable changes in mitochondrial morphology were observed in the peripodial membrane, likely because the duration of transgene expression was insufficient to elicit phenotypic alterations in this specific tissue. Therefore, while we aimed to compare this genotype to Gug>dMfn and Gug>miro, the technical limitations prevented a conclusive analysis.
We have prepared a representative figure for the reviewer (below), showing representative confocal images of wing discs showing mito-GFP and Dapi in the three genotypes indicated in the Fig.
In Fig. 3I: what is really the mitochondrion? It would be good to outline the region(s) that was/were measured.
To improve clarity, we have repeated the electron microscopy (EM) analysis and now provide representative images that more clearly illustrate mitochondrial morphology in the different genotypes analyzed. These updated images presented in Fig 4 better highlight the structural alterations observed upon genetic manipulation and help clarify the basis for our morphological assessments.
We have extended our analysis and have assessed mitochondrial ultrastructure in Yki + dPGC1-RNAi wing disc tumors, with and without dMfn1 downregulation—the most upregulated mitochondrial fusion gene in this tumor context. In Yki + dPGC1-RNAi tumors, mitochondria appeared more elongated, consistent with increased fusion. Upon dMfn1 depletion, we observed a dramatic shift in mitochondrial morphology: mitochondria became larger and more rounded, with disrupted cristae and onion-like structures, indicative of compromised mitochondrial integrity and function (see new Fig 4).
A quantification of RNAi and overexpression efficiencies of the different transgenes in Fig. 3 is required.
To assess the efficiency of RNAi-mediated knockdown and transgene overexpression, we performed quantitative PCR (qPCR) using the ubiquitous Actin-Gal4 driver. While we acknowledge that this driver does not replicate the spatial specificity of the periodic membrane Gal4 driver used in the experiments shown in Figure 3 (Gug-Gal4), the latter targets a very limited number of cells within the imaginal disc, making reliable qPCR quantification unfeasible.
Using Actin-Gal4 allows us to obtain a relative and informative measure of transgene efficiency across the different constructs. These data confirm effective knockdown and overexpression of the relevant genes and are now included in Figure S2.
In Fig. 4: what is the phenotype when miro is over-expressed in combination with Yki? Or when it is knocked down in the ap>Yki-dPGC1 background? This was the gene tested in Fig. 3 with a clear mitochondrial phenotype
To address whether miro contributes to Yki-mediated tumor growth, we performed the requested experiments and now include the results in the revised manuscript (see updated Results section, lines 374-377, and new Fig. S11).
Our data show that overexpression of miro in combination with Yki does not lead to a significant increase in tissue growth or tumor-like phenotypes, in contrast to the effects observed with dMfn or Opa1 overexpression. Similarly, knockdown of miro in the ap>Yki-dPGC1-RNAi background did not suppress tumor growth, indicating that miro is not required for the enhanced proliferation observed in this context.
These findings suggest that, although miro influences mitochondrial morphology in normal wing discs (as shown in Fig. 3), its role in tumorigenesis is distinct from that of dMfn and Opa1. We have revised the manuscript to clarify the gene-specific contributions of mitochondrial fusion regulators to Yki-driven tumorigenesis. This distinction underscores the complexity of mitochondrial dynamics and highlights that not all fusion-related genes exert the same functional impact in oncogenic settings.
How does the mitochondrial morphology in the wing disc peripodial epithelium look like in Gug>Opa1RNAi or Gug>Opa1 discs?
To assess the impact of Opa1 on mitochondrial morphology in the peripodial epithelium of the wing disc, we used the Gug-GAL4 driver to either overexpress or knock down Opa1. Our analysis revealed that Opa1 overexpression led to slightly elongated mitochondria, but did not result in extensive network formation, suggesting a modest enhancement of inner membrane fusion. In contrast, Opa1 knockdown caused clear mitochondrial fragmentation, closely resembling the phenotype observed upon dMfn depletion. These results shown in Fig 3 are consistent with the distinct roles of Opa1 and dMfn in regulating mitochondrial fusion: Opa1 primarily modulates inner membrane fusion and cristae architecture, while dMfn drives outer membrane fusion and network connectivity.
The corresponding data are presented in Figure 3F, G, and quantified in Figure S9, alongside experiments manipulating other genes involved in mitochondrial dynamics.
Why have the authors switched between the ap>Yki+dPGCRNAi and the ap>Yki+dPGC1shRNA lines? It would be important to have this series of experiments in the same backgrounds, as KD efficiencies are different (Fig. S1C).
The primary reason for switching between the dPGC1-RNAi and dPGC1-shRNA lines was practical: the chromosomal insertion sites of the transgenes made certain genetic combinations more feasible with one line over the other. This flexibility significantly facilitated our experimental design and analysis.
To address concerns regarding knockdown efficiency, we performed a comparative analysis using the ubiquitous actin-GAL4 driver, rather than MS1096-GAL4, which exhibits patchy and dynamic expression in the wing imaginal disc. This allowed us to obtain a more consistent and interpretable measure of mRNA downregulation for both transgenes. Our results show that both lines achieve comparable levels of knockdown, as shown in Figure S2.
Fig. 5A: proper quantification of Western Blot signals is required. I do not agree that Cyclin E protein levels are elevated in ap>Yki or ap>Yki+dPGC1 discs. Even at the mRNA levels the increase in expression is rather weak. From these results nothing can be concluded.
We have repeated the Western blot analysis using seven independent membranes to ensure robust quantification of Cyclin E levels in ap>Yki and ap>Yki+dPGC1-RNAi wing discs (Fig 6).
Although the increase in Cyclin E protein levels is subtle, it is consistent across replicates and statistically significant. We have now included the quantification of these Western blot signals in the revised Figure 6, which supports the conclusion that Cyclin E levels are elevated in ap>Yki+dPGC1 discs.
We hope this additional data addresses the reviewer’s concern and strengthens the interpretation of our results.
Knock-down efficiencies for dap and CycE needs to be quantifiec (Fig. 5H-N). Although the rescue experiment with CycE knock down is from the phenotype convincing, it is nonetheless puzzling, as CycE is accodring to Fig. 5A+B hardly upregulated. An independent CycE RNAi line would be useful.
We have quantified the knockdown efficiency of the dap-RNAi line, and the results are included in Figure S13.
Regarding Cyclin E, we would like to clarify that we did not use an RNAi line in this experiment. Instead, we employed the CycE-05306 mutant allele in a heterozygous background, which is expected to reduce Cyclin E levels by approximately 50%. The CycE-05306 allele in Drosophila melanogaster is a loss-of-function allele of the Cyclin E gene. This allele carries a P-element insertion in the first intron of the CycE gene, which disrupts normal transcription and reduces Cyclin E expression. In a heterozygous background, as used in your experiments, CycE-05306/+ is expected to reduce Cyclin E levels by approximately 50%, which is typically sufficient to observe genetic interactions or sensitized phenotypes without affecting normal development. This makes it a valuable tool for studying gene dosage effects, particularly in tumor models where Cyclin E activity may be rate-limiting.
Importantly, this partial reduction does not impair normal tissue growth, but it strongly limits tumor growth in the context of Yki overexpression combined with dPGC1 downregulation, as shown in Figure 6. This selective sensitivity highlights the functional importance of Cyclin E in supporting oncogenic growth driven by Yki and dPGC1 depletion. We believe this provides compelling evidence for Cyclin E’s role in this tumor model.
Reviewer #3 (Significance (Required)):
Strengths and Limitations of the Study Strengths Innovative Focus on Mitochondrial Dynamics and Oncogenesis: The study provides compelling evidence linking mitochondrial dynamics, particularly hyperfusion, to tumorigenesis in Drosophila. The identification of dPGC1 as a context-dependent tumor suppressor adds novel insights into the interplay between metabolism and oncogenesis. Comprehensive Use of Drosophila as a Model System: The study leverages the genetic tractability of Drosophila, allowing precise manipulation of mitochondrial regulators and signaling pathways. The use of wing imaginal discs as a model for tumor growth is well-established and appropriate. Integration of Morphological and Genetic Data: The manuscript combines confocal imaging, electron microscopy, and genetic tools to demonstrate the role of dPGC1 in regulating mitochondrial dynamics, Cyclin E levels, and tissue overgrowth. Relevance to Cancer Biology: The findings address key hallmarks of cancer, including deregulated metabolism, genomic instability, and cell cycle misregulation. The study's exploration of these processes in a simple model organism provides a strong basis for translating findings to mammalian systems.
Limitations Validation of RNAi and Overexpression Efficiency: The knockdown efficiency of dPGC1 on the mRNA level is only moderate (30-50%), and protein-level validation is missing. Without this, the study cannot conclusively demonstrate the role of dPGC1 in normal development or tumorigenesis. Incomplete Mechanistic Insights: The manuscript identifies Cyclin E as a potential driver of tumor growth but does not adequately explore how mitochondrial hyperfusion leads to Cyclin E regulation (e.g., post-transcriptional mechanisms or protein stability). Inconsistencies in Experimental Backgrounds: The study uses different RNAi/shRNA lines and driver combinations inconsistently across experiments, making it difficult to compare results directly. This variability undermines the robustness of the conclusions. Limited Functional Analysis of Mitochondria: While mitochondrial morphology is well-characterized, functional assays (e.g., membrane potential or ATP production) are missing. These would confirm the impact of hyperfusion on cellular energetics and oncogenesis.
In the revised manuscript, we have addressed each of the concerns raised.
In addition to that, in the revised version of the manuscript, we have included new experiments to assess mitochondrial functionality in tumors co-expressing Yki and dPGC1-RNAi. Specifically, we analyzed the Mitochondrial Membrane Potential (MMP). We used TMRE staining to evaluate MMP, a key indicator of mitochondrial integrity and oxidative phosphorylation capacity. Our analysis revealed no significant differences in MMP between Yki tumors and Yki + dPGC1-RNAi tumors, suggesting that mitochondrial membrane potential is preserved despite the observed morphological abnormalities. These results are shown in Fig S6. In the text it is discussed in lines 233-243.
Contribution to Existing Literature The study makes a significant contribution to the growing body of literature on the metabolic regulation of cancer by identifying dPGC1 as a tumor suppressor modulating mitochondrial dynamics. Previous work has established the dual roles of mammalian PGC1α in promoting or suppressing cancer depending on context. This study adds depth by demonstrating similar context-dependent effects in a simpler model organism, facilitating further exploration of the molecular mechanisms involved.
By linking mitochondrial fusion, Yki signaling, and Cyclin E regulation, the manuscript aligns with and expands upon research on Hippo pathway regulation, cancer metabolism, and mitochondrial biology. The findings highlight the importance of integrating metabolic and signaling networks in understanding oncogenesis.
Community Selection The current form of the manuscript is best suited for a specialized audience, particularly mitochondrial biologists, Drosophila researchers, and Hippo pathway specialists. To engage a broader community, additional work linking these findings to mammalian models or human cancer biology would be necessary.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript presents compelling evidence that dPGC1 acts as a context-dependent tumor suppressor in Drosophila by modulating mitochondrial dynamics and limiting Yorkie (Yki)-induced oncogenic growth. By leveraging the Drosophila wing imaginal disc as a model, the authors investigate how dPGC1 depletion exacerbates Yki-driven tissue overgrowth, mitochondrial hyperfusion, Cyclin E upregulation, and DNA damage, leading to tumorigenesis. The study provides valuable insights into the interplay between mitochondrial dynamics and cancer, with implications for understanding metabolic regulation in oncogenesis. While the findings are significant and well-aligned with the field, certain aspects of the experimental design, data presentation, and mechanistic insights require further attention to enhance clarity, reproducibility, and impact. Below, I outline my major concerns and recommendations.
Major Points
Strengths and Limitations of the Study
Strengths
Innovative Focus on Mitochondrial Dynamics and Oncogenesis: The study provides compelling evidence linking mitochondrial dynamics, particularly hyperfusion, to tumorigenesis in Drosophila. The identification of dPGC1 as a context-dependent tumor suppressor adds novel insights into the interplay between metabolism and oncogenesis. Comprehensive Use of Drosophila as a Model System: The study leverages the genetic tractability of Drosophila, allowing precise manipulation of mitochondrial regulators and signaling pathways. The use of wing imaginal discs as a model for tumor growth is well-established and appropriate. Integration of Morphological and Genetic Data: The manuscript combines confocal imaging, electron microscopy, and genetic tools to demonstrate the role of dPGC1 in regulating mitochondrial dynamics, Cyclin E levels, and tissue overgrowth. Relevance to Cancer Biology: The findings address key hallmarks of cancer, including deregulated metabolism, genomic instability, and cell cycle misregulation. The study's exploration of these processes in a simple model organism provides a strong basis for translating findings to mammalian systems.
Limitations
Validation of RNAi and Overexpression Efficiency: The knockdown efficiency of dPGC1 on the mRNA level is only moderate (30-50%), and protein-level validation is missing. Without this, the study cannot conclusively demonstrate the role of dPGC1 in normal development or tumorigenesis. Incomplete Mechanistic Insights: The manuscript identifies Cyclin E as a potential driver of tumor growth but does not adequately explore how mitochondrial hyperfusion leads to Cyclin E regulation (e.g., post-transcriptional mechanisms or protein stability). Inconsistencies in Experimental Backgrounds: The study uses different RNAi/shRNA lines and driver combinations inconsistently across experiments, making it difficult to compare results directly. This variability undermines the robustness of the conclusions. Limited Functional Analysis of Mitochondria: While mitochondrial morphology is well-characterized, functional assays (e.g., membrane potential or ATP production) are missing. These would confirm the impact of hyperfusion on cellular energetics and oncogenesis.
Contribution to Existing Literature
The study makes a significant contribution to the growing body of literature on the metabolic regulation of cancer by identifying dPGC1 as a tumor suppressor modulating mitochondrial dynamics. Previous work has established the dual roles of mammalian PGC1α in promoting or suppressing cancer depending on context. This study adds depth by demonstrating similar context-dependent effects in a simpler model organism, facilitating further exploration of the molecular mechanisms involved.
By linking mitochondrial fusion, Yki signaling, and Cyclin E regulation, the manuscript aligns with and expands upon research on Hippo pathway regulation, cancer metabolism, and mitochondrial biology. The findings highlight the importance of integrating metabolic and signaling networks in understanding oncogenesis.
Community Selection
The current form of the manuscript is best suited for a specialized audience, particularly mitochondrial biologists, Drosophila researchers, and Hippo pathway specialists. To engage a broader community, additional work linking these findings to mammalian models or human cancer biology would be necessary.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary
In this manuscript the authors the investigate the role of the mitochondrial regulatory transcription factor dPGC1 in tissue growth and oncogenic transformation. They show that dPGC1 limits hyperplasia mediated by overexpression of Yki in the Drosophila wing disc, while having no effect on normal growth. dPGC1 depletion in discs overexpressing Yki results neoplastic overgrowth and hyperfused mitochondria, which was dependent on the increased expression of genes involved in promoting mitochondrial fusion. Additionally, the authors show that dPGC1 limits CycE levels post-transcriptionally in Yki tumors.
Major comments
Minor Comments:
The manuscript by Sew et al builds on the previous work by Nagaraj et al to explore the role of mitochondrial function in tumors driven by disruption of the Hippo pathway. In particular, the authors identify dPGC1 as a transcription factor that limits Yki-driven mitochondrial fusion and tissue growth. Interestingly, they further show that Yki/PGC1-depleted tumors are highly sensitive to Cyclin E levels, due to post-transcriptional Cyclin E increase. These results further our knowledge of how Yki drives growth and how mitochondria participate in oncogenic transformation. With appropriate revision as outlined above (for example exploring whether the mechanism proposed is Yki-specific), the manuscript will be of broad interest to developmental and cancer biologists.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Sew et al. examine the master regulator of mitochondrial biogenesis, dPGC1, in the context of Drosophila wing and larval development. They primarily use confocal imaging to probe the interplay between dPGC1 and an overactive Hippo pathway, driven by overexpression of the main effector protein, Yki. In their study, they find that tumors, driven by overactivity of Yki grow larger when dPGC1 is downregulated, implicating the mitochondrial biogenesis pathway in tumor suppression. Furthermore, in the context of Yki overexpression, they find that levels of Mfn or Opa1 modulate tumor size. Lastly, they show a role of cyclin E in controlling the size of tumors formed by Yki OE + dPGC1 RNAi. The potential role of dPGC1 as a tumor suppressor is interesting because it highlights an emerging recognition of mitochondria in the aetiology of cancer. However, before publication, much of the data in this manuscript should be strengthened by a refinement in the methods/analysis and an increase in orthogonal approaches.
Major comments:
The authors indicate that for example, in lines127-28, that neither downregulating or overexpressing dPGC1 affects wing size. However, the quantification in Fig. 1C shows a significant decrease in wing size following RNAi treatment. This decrease is modest, but it is nevertheless significant. It is worth pointing out, too, that the efficiency of the RNAi in Fig. S1C suggests that the conclusions drawn are premature. While a roughly 55% drop in mRNA levels may be statistically significant, it is unclear whether this drop in transcripts corresponds to a commensurate depletion of protein. Moreover, it is unclear, in this context, how much dPGC1 may indeed be necessary to drive a relatively normal program of mitochondrial biogenesis in wing development. To obtain a clear result, it is necessary to show significant depletion of the dPGC1 protein. (Ultimately, if it is the case that dPGC1 is unnecessary for wing development and function, a more coherent line of inquiry would be to find out the reason for this rather than to pivot the story to studying tumorigenesis in larva.)
In Figure 3H-K, it is not clear why the authors used electron microscopy to evaluate mitochondrial morphology. The very good confocal images in Figure 3C-G show a clear change in mitochondrial morphology following the knockdown of Mfn, Opa1, and Miro. While it is clear from the electron micrographs in Figure H that the mitochondria are enlarged, it is not obvious that this increase in length is a result of increased mitochondrial fusion. Indeed, if the mean form factor were used to quantify the shape, it is likely that in both conditions, the value would be close to 1, indicating more of a round object, and it not obvious whether there would be a difference between the Yki OE versus the YkI OE + dPGC1 RNAi. Therefore, from this data alone, it cannot be concluded that the YkI OE + dPGC1 RNAi condition leads to mitochondrial hyperfusion.
Figure 4. refers to changes in mitochondrial fusion and fission in tumor formation; however, the authors do not attempt to alter mitochondrial fission factors, so it is not accurate to mention a role of mitochondrial fission, in this context. It must be noted, too, that the authors have not demonstrated that their genetic interventions have actually affected mitochondrial morphology in these experiments. As noted in the previous figure, the Yki OE + dPGC1 RNAi condition showed enlarged mitochondria, but not necessarily hyperfused organelles. Therefore, the downregulation of Mfn or Opa1 in this set of experiments may not necessarily have altered mitochondrial morphology. Perhaps suppression of Mfn or Opa1 would normalize the areas of these evidently swollen mitochondria, but this is unclear without images. Furthermore, it should be appreciated that both Opa1 and Mfn exhibit pleiotropic attributes - e.g., Opa1 not only regulates IMM fusion, but it also modulates the shape and tightness of cristae membranes, specialized sites of oxidative phosphorylation as well as sequestration of cytochrome c, the release of which influences apoptosis (Frezza et al., 2006). At least in mammalian cells, Mfn2 is thought to regulate contacts between mitochondria and endoplasmic reticulum (Naon et al., 2023), which may serve other functions than OMM fusion, such as stabilization of the MAM.
Figure 5 highlights a connection between dysregulation of mitochondria and Cyclin E, which allows cells to prematurely enter S phase. The data presented here do not offer clarity on whether the enlargement of the tumors results from increase cellular proliferation and/or cell size. The role of the cell cycle adds a layer of complexity to these results, because it is thought that mitochondria undergo fragmentation during the cell cycle to promote an even distribution of the organelle population after mitosis (Taguchi et al., 2007); however, in this manuscript, the authors contend that the downregulation dPGC1 is promoting mitochondrial hyperfusion. It is unclear how and whether cellular division and proliferation would proceed at an accelerated rate in a situation with mitochondrial hyperfusion.
Minor comments:
Lines 69-72 contrast the roles of PGC1α and β. It is not clear whether the comparison is of their respective roles in cancer or in normal physiology. In either case, it is important to note that PGC1β has been shown to drive mitochondrial fusion as well as biogenesis through its control of MFN2, among other factors (Liesa et al., 2008).
Although this study focuses on PGC1, the authors do not seem to site the original literature from the Spiegelman lab.
There are 10-20 grammatical errors throughout the text.
Referee Cross-commenting
There is agreement among the referees that the potential role of PGC1 as a tumor suppressor is interesting and significant. However, various aspects of this work require attention prior to publication. For example, there needs to be a complete knock down of PGC1 to come to any conclusion as to its role in wing development. The methods for analyzing mitochondrial morphology need to be clarified and be consistent with standards in the field of mitochondrial dynamics. Also, the authors need to quantify their Western blots to obtain accurate assessments of protein levels. Generally, the study relies too heavily on overexpression experiments; understanding the potential role of mitochondria in regulating the Hippo pathway should include various knockdown and/or knockout models.
Overall, the authors show an interesting dampening effect of dPGC1 on growth of Yki-driven tumors. This data could be relevant for elucidating how dysregulation of the Hippo signalling pathway can underlie tumorigenesis.
The narrative arc of the study, however, appears to lack a focused line of inquiry. Figure 1 highlights an attempt to modulate Drosophila wing size and/or structure by downregulating dPGC1, but to no effect. Although examination of the efficiency of the RNAi revealed that the transcripts were still present in significant quantities; so, the conclusion that dPGC1 is dispensable for wing formation is premature. To have clarity on this point, it would be necessary to completely knockdown the gene, preferably by showing a total loss of protein. This should be feasible for the authors, since they showed Western blotting in Figure 5A. In any event, it seems that this negative data led the authors to study the Hippo pathway in the larval stage. This transition from Figure 1 to 2 seemed somewhat arbitrary and leads to a rather disjointed sense of the main line of inquiry around dPGC1.
It is important to note, too, that the authors highlight a role of mitochondrial dynamics in the pathway of Yki-driven tumor formation; however, they only directly evaluate mitochondrial dynamics in this context in a single assay, namely, Figure 3H-K, and this quantification is likely inaccurate because the mitochondria in the Yki OE + dPGC1 RNAi condition seem to be substantially enlarged, circular structures. It is critical to keep in mind that mitochondrial enlargement does not necessarily stem from hyperfusion. It could come from a decrease in the activity of Drp1 or result from an imbalance between mitochondrial biogenesis and mitophagy.
A marked limitation of this study is the overuse of rather artificial manipulations of transcriptional regulatory pathways. The study would benefit a lot from investigation of the loss of function of components of the Hippo pathway rather than just OE of Yki.
My expertise is in mitochondrial biology, with specialization in super-resolution imaging, mitochondrial dynamics and membrane architecture. I have also worked in the interface between mitochondrial physiology and cancer. With this perspective, I think that the authors uncover a potentially interesting role of PGC1 as a tumor suppressor.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Manuscript number: RC- 2025-03073
Corresponding author(s): Shaul Yogev
We kindly thank our reviewers for their enthusiasm, thoughtful feedback, and constructive suggestions on how to strengthen our manuscript. Below, we provide a point-by-point response to reviewer comments and outline the experiments we will do to address every concern that has been raised.
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
This interesting study uses an unbiased genetic screen in C. elegans to identify SAX-1/NDR kinase as a regulator of dendritic branch elimination. Loss of SAX-1 results in an excess branching phenotype that is striking and highly penetrant. The authors identify several additional regulators of branch elimination (SAX-2, MOB-1, RABI-1, RAB-11.2) by using a candidate genetic screen aimed at factors that interact physically or genetically with SAX-1. They propose that SAX-1 acts by promoting membrane retrieval based on the nature of these interactors and the results of an imaging-based in vivo assay for endocytic puncta.
Major comments.
To me, the simplest genetic explanation is that daf-7 and daf-2 are partially required for branch retraction in a manner redundant with sax-1, and the ts mutants are not fully wild-type at 15C. Thus, the sax-1 requirement is revealed only in these mutant backgrounds. Can the authors examine starvation-induced dauers of daf-7 or daf-2 raised continuously at 15C?
We will do this experiment.
daf-7 and daf-2 ts strains can form "partial dauers" that have a dauer-like appearance but are not SDS resistant. Could the difference between partial dauers and full dauers account for the difference in sax-1-dependence? The authors could use SDS selection of the daf-7 strain at 25C to ensure they are examining full dauers.
We tested daf-7 mutants with 1% SDS when we set up the system – they are fully dauer at 25°C and are SDS sensitive after exit. We will repeat this important control with daf-7; sax-1 double mutants.
The Bargmann lab has created a daf-2 FLP-OUT strain (ky1095ky1087) that allows cell-type-specific removal of daf-2. Could this be used to test for a cell-autonomous role of daf-2 in IL2Q related to branch elimination?
We can attempt this experiment. However, since IL2 promoters turn on prior to dauer, the interpretation would not be straightforward – it would be hard to exclude that a cell autonomous defect in dauer entry does not account for the IL2 dauer exit phenotype, even if branching appears normal.
These ideas are not a list of specific experiments the authors need to complete, rather they are meant to illustrate some possible approaches to the question. Whatever approach they use, it is important for them to more rigorously explain why SAX-1 is not required for branch removal in wild-type animals.
We completely agree. We will carry out the 15°C experiment, examine morphological characteristics and test SDS resistance. In addition, we will test neuronal markers that differ between dauers and non-dauers to determine whether the mutants are full or partial dauers at the relevant timepoints.
The SAX-2 localization (Fig. 4) and endocytosis assay (Fig. 6) results were not clear to me from the data shown. Overall a more rigorous analysis and presentation of the data would be important to make these conclusions convincing. This may involve refining the data presentation in the figures, modifying the claims (e.g., "we propose" vs "we find"), or saving some of the data to be more fully explored in a future paper. In my view, these figures are the biggest weak point of the manuscript and also are not important for the central conclusions (which are well supported and convincing), indeed these results are barely mentioned in the Abstract or last paragraph of Introduction.
We agree that the analysis and presentation of Figures 4 and 6 need to be improved. The presentation has already been updated, and the figures are clearer now. In the revision, we will increase sample size to provide stronger conclusions, consolidate some of the analysis and further improve presentation. While we agree with the reviewer that conclusions from these figures are not as strong as those drawn from genetic experiments, they do complement and support the conclusions of those other figures.
There is no bleed-through: this is most evident by looking at the brightest signals in the cell body (now labelled with an asterisk in a zoomed-out image) and noting that they do not bleed between channels. In sax-1 mutants, the SAX-2::GFP puncta are very obvious and distinguishable from the tagRFP channel. In control, SAX-2::GFP is very faint in the dendrite, so we increased the contrast to allow visualization. The reviewer is correct that under these conditions, some puncta look like the cytosolic fill. In the revision, we will re-analyze the data and will not consider these as bona-fide SAX-2 puncta, but rather cytosolic SAX-2 that accumulates due to constrictions and varicosities in the dendrite.
We generated an endogenously tagged sax-1 with a 7xspGFP11 tag; however, this was below detection in the IL2s. For the revisions, we can test an overexpressed cDNA construct.
**Referee cross-commenting**
I think we all touched on similar points. I wanted to follow up on Reviewer 3's comment, "Is the failure to eliminate branches an indication of incomplete dauer recovery? Do sax-1 mutants retain additional characteristics of dauer morphology in post dauer adults." I thought this was an excellent point. It made me wonder if that might explain why the defect is only seen in daf-7 and daf-2 mutant backgrounds - maybe these strains retain partial dauer traits even after exit. Is there a specific experiment that they could do? Did you have specific characteristics of dauer morphology in mind for them to check? (Ideally something in the nervous system that can be scored quantitatively.)
Please see response to point #1 regarding experiments we will do to confirm the “dauer state” of daf-7 and daf-7; sax-1 double mutants.
Reviewer #1 (Significance (Required)):
A major strength of this work is the pioneering use of a novel system to study neuronal branch retraction. C. elegans has provided a powerful model for studying how dendrite branches form, but much less attention has been paid to how excess neuronal branches are removed. The post-dauer remodeling of IL2Q neurons provides an exciting and dramatic physiological example to explore this question.
This paper is notable for taking the first steps towards developing this innovative model. It does exactly what is needed at the outset of a new exploration - a forward genetic screen to discover the main regulators of the process. Using a combination of classical and modern genetic approaches, the authors bootstrap their way to a sizeable list of factors and a solid understanding of the properties of this system, for example that retraction of higher vs lower order dendrites show different genetic requirements.
We thank the reviewer for recognizing the novelty and significance of our work.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
In this manuscript, the authors establish C. elegans IL2 neurons as a system in which to study dendrite pruning. They use the system to perform a genetic screen for pruning regulators and find an allele of sax-1. Unexpectedly sax-1 is only required for post-dauer pruning in two different genetic backgrounds that induce dauer formation, but not starvation-induced dauer formation. Sax-1/NDR kinase reduction has previously been associated with increased outgrowth and branching in other systems, so this is a new role for this protein. However, the authors show that proteins that work with Sax-1 in other systems, like sax-2/fry, also play a role in this pathway. The genetic experiments are beautiful and the findings are all clearly explained and strongly supported. The authors also examine sax-2 localization, which localizes sax-1 in other systems, and show it in puncta in dendrites that increase with dauer exit, consistent with function at the time of pruning. They also show that membrane trafficking regulators associated with NDR kinases function in the same pathway here, hinting that endocytosis may play a role during pruning as in Drosophila. The link to endocytosis was a little weak (see Major point below). Overall, this study describes a new system to study pruning and identifies NDR/fry/Rabs as regulators of pruning during dauer exit. The work is very high quality and both the imaging and genetics are extremely well done.
We thank the reviewer for their positive assessment of the manuscript.
Major points
Reviewer #2 (Significance (Required)):
Neurite pruning is important in all animals with neurons. Genetic approaches have primarily been applied to the problem using Drosophila, so identifying a new model system in which to study it is an important step. Using this system, a pathway known to function in a different context is linked to pruning. Thus the study provides new insights into both pruning and this pathway.
We thank the reviewer for the positive assessment of our study’s significance.
__Reviewer #3 (Evidence, reproducibility and clarity (Required)): __
Summary: Figueroa-Delgado et al. use a C. elegans neuro plasticity model to examine how dendrites are eliminated upon recovery from the stress induced larval stage, dauer. The authors performed a mutagenesis screen to identify novel regulators of dendrite elimination and revealed some surprising results. Branch elimination mechanism varies between 2{degree sign}, 3{degree sign}, and 4{degree sign} branches. The NDR kinase, SAX-1 and it's interactors (SAX-2 and MOB-2) are required for elimination of second and third order branches but not fourth order branches. Interestingly they showed that branch elimination varies depending on the stimulus of dendrite outgrowth such that the NDR kinase is required for branch elimination after genetically inducing the dauer stage but is not required if dauers are produced through food deprivation. The authors go a step further to include a small candidate screen looking at various pathways of membrane remodeling and identify additional regulators of dendrite elimination related to membrane trafficking including RABI-1, RAB-8, RAB-10, and RAB-11.2.
We thank the reviewer for their time and suggestions below
Major comments:
While I find the data promising and exciting, several of the experiments have concerningly low sample sizes. Fig 3G, Fig 4G, Fig 5J and L, and Fig 6I all contain data sets that are fewer than 10 animals. Sample sizes should be stated specifically in the figure legends for all data represented in the graphs. We thank the reviewer for finding the data exciting. We agree that the sample sizes in some panels is low and will increase it in the revised version. Sample sizes are now specifically listed in the figure legends.
All statements based on data not shown should be amended to include the data as a supplemental figure or edited to omit the statement based on withheld data. We agree. Some “not shown” data are already added to the current version of the manuscript and the rest will be added to the fully revised version, or the statements will be omitted.
Rescue experiments (Fig 2J) should demonstrate failure to rescue from neighboring tissue types (hypodermis and muscle) to conclude cell autonomous rescue rather than a broadly acting factor. Thank you for the suggestion. We will use a hypodermal promoter and a muscle promoter driving SAX-1 cDNA expression to strengthen the claim of cell autonomy.
Fig 4 needs quantification of higher order branches and SAX-2 proximity to branch nodes as these are discussed in the text. We will add this quantification.
Minor comments:
Fig 1C-F, It appears like the shy87 allele produces animals of significantly different body sizes. It would improve rigor to normalize the dendrite coverage to body size in the quantification. We do not see a biologically meaningful size difference between shy87 and control, it may be the specific image shown. We will confirm this by measuring animal size for the final revision.
Is the failure to eliminate branches an indication of incomplete dauer recovery? Do sax-1 mutants retain additional characteristics of dauer morphology in post dauer adults. This important point was also raised by Reviewer 1. We will test SDS sensitivity, morphological markers, and molecular markers to determine the dauer “state” of the mutants used in this study. The results will be included in the final revision.
The text references multiple transgenic lines tested in Fig 2I-J but only one line is shown. Additional lines were visually examined under a fluorescent compound microscope but not imaged or quantified. We will add this quantification to the final revision.
Fig 4F, Additional timepoints would enhance the sax-1 localization result and might provide insight into mechanism of action for sax-1. We will add the localization in post-dauer adults.
Fig 6I Control and sax-1(ky491) example images should be provided in the supplement. We will add these images to the final revision.
**Referee cross-commenting**
I agree that we shared many of the same concerns.
There are several general assays for dauer characteristics that could be used here to determine if the post-dauer animals retain other characteristics of the dauer stage in addition to IL2 branches (SDS resistance, alae remodeling, pharyngeal bulb morphology, nictation behavior). The nictation behavior has been connected very nicely with IL2 neurons (Junho Lee's group). Additionally, FLP dendrites occupy the same space as the IL2 branches and outgrowth in post-dauers occurs in coordination with IL2 branch elimination - this might be another optional experiment, to check if FLP growth is impeded by persistent IL2 branches. All of these could be quantified similar to how the authors have already established with their IL2 model (FLP dendrite branches) or with a binary statistic.
Please see responses to Reviewer 1 and 3 above for the list of experiments to determine whether the animals fail to completely enter or exit dauer.
Reviewer #3 (Significance (Required)):
SIGNIFICANCE ============ These results describe a new role for the NDR kinase complex in dendrite pruning that has clinical significance to our understanding of human brain development and human health concerns in which pruning is dysregulated, such as observed in the case of autism. The authors use an established neuro-plasticity, C. elegans model (Schroeder et al. 2013) which provides a tractable and reproduceable platform for discovering the mechanism of dendrite pruning. These results would influence future work in the fields of cell biology of the neuron and disease models of brain development.
My expertise is in the field of C. elegans neuroscience and stress biology and have sufficient expertise to evaluate all aspects of this work.
Reviewer #1
We apologize for a mistake in the arrowhead color and overall presentation of this figure. It has been fixed in the current version.
We thank Reviewer #1 for their observation, and we apologize for our oversight. We fixed this in the current version.
We added zoomed-out images and indicated where the zoomed in insets are taken from. We thank the reviewer for helping us improve the clarity of the data.
This is a very good point. The increase in SAX-2 puncta in sax-1 mutants is stronger during dauer-exit than in dauer, consistent with this being the time when SAX-1 functions. We agree that some earlier activity of SAX-1 cannot be excluded, and we do not assume that the effect on SAX-2 completely accounts for the pruning defects. This is now acknowledged in the text. However, given that both proteins function together in pruning, and given that the effect is strongest during dauer exit, we do believe that this data is informative and worth showing.
There is no correlation. In other words, the number of SAX-2 puncta does not instruct the extent of pruning. Please note the quantifications underestimate the number of SAX-2 puncta in the mutants, since they were only done on the primary dendrite. This is necessary because the mutant and control have different arbor size, so only branch order that can be appropriately compared are primary dendrites.
We thank the reviewer for raising this point and apologize for the oversight in data presentation. In the revised manuscript, we now show all control and experimental data integrated into a single graph, ensuring that each dataset is represented accurately to provide a comparison between dauer and post dauer recovery conditions.
We sincerely apologize for this mistake, some of the data was erroneously grouped in the original submission. The revised version contains an updated number of neurons, presented on the same graph, and in the final revision we will further increase sample size. We apologize again for this error.
Based on EM, both an endocytic punctum and the diameter of the neuron are smaller than a single pixel. The apparent difference in size in fluorescence microscopy is because the puncta are brighter (they contain more membrane) and thus appear larger. In the current version, the improved presentation of the figure contains zoomed out images that clearly show that there is no bleed-through.
CD8 lacks clear endocytosis motifs, which is why it is advantageous for labelling neurites and testing endocytosis when paired with an endocytic signal (Lee and Luo 1999; Kozik et al. 2010). Conversely, extracellular GFP binding to a membrane GFP antibody can induce endocytosis (for example, see (Tang et al., 2020)), likely by inducing clustering, although we are not familiar with work that explored the mechanism. In the updated version we included a rare example of an mCD8 punctum.
We apologize for the presentation in the original version of Figure 6. This impression was because we showed single focal planes that only captured some of the signal. In the revised version we show projections, which makes it evident that there are fewer endocytic events in the mutant.
These puncta are secreted or muscle-associated GFP that has not been internalized by IL2Q neurons. They are present in all strains in this figure, this can be clearly seen in the zoomed-out images that have been added to the updated figure.
This is indeed the soma. In the updated version this can be clearly seen in the zoom-out. The large puncta in the soma were not counted because they may arise from the fusion of an unknown number of smaller puncta, and their precise number cannot be determined at the resolution of fluorescence microscopy.
We thank the reviewer for catching this oversight, it is now fixed.
Minor points:
In Fig. 1A, C. elegans is shown going directly from L1 to dauer in response to unfavorable conditions, which is incorrect. Animals proceed through L2 (in many cases actually an alternative L2d pre-dauer) and then molt into dauer (an alternative L3 stage) after completing L2.
We updated the schematic to include the L2d stage where commitment to dauer entry or resumption to reproductive development is made.
In Fig. 1B, please check if it is correct that hypodermis contacts the pharynx basement membrane as drawn. The schematic in the top panel makes it look like there is a single secondary branch and the quaternary branches are similar in length to the primary dendrite. The schematic in the bottom panel makes it look like the entire neuron is a small fraction of the length of the pharynx. Could these be drawn closer to scale?
The hypodermis does contact the pharynx basement membrane. We redrew the schematic for clarity.
Reviewer #2
For context, it might be helpful to know whether branching of other dendrites is increased in sax-1 mutants (as expected based on phenotypes in other animals) or decreased like IL2 neurons.
We examined the branching pattern of PVD, a polymodal nociceptive neuron (new Supplemental Figure 3). We find no significant difference between control and sax-1 or sax-2 mutants, suggesting that these genes function in the context of pruning. Recent work (Zhao et al. 2022) confirms that sax-1 is not required for PVD branching.
Minor:
"shy87 mutant dauers showed a minor reduction in secondary and tertiary branches compared to control (Figure 1G). These results indicate that shy87 is specifically required for the elimination of dauer-generated dendrite branches." Maybe temper the specificity claim some as the reduction in branches is definitely there.
We agree, the claim was tempered.
"three complimentary approaches" should be complementary
Thank you for noticing. We fixed this.
"In control animals, SAX-2 was mostly concentrated in the cell body (data not shown)" It might be nice to include some overview images that show the cell body for completeness.
We added zoomed-out images to the revised figure, thank you for the suggestion.
Reviewer #3
Minor comments:
Fig 1G-H, are shy87 second and third order branch counts statistically different between dauer and post dauer adults? This comparison would strengthen the claim that these order branches fail to eliminate all together rather than undergo a partial elimination. We added this to Figure S2. The shy87 mutants show a complete failure in eliminating secondary branches (i.e. no difference between dauer and post-dauer) and a strong but incomplete defect in eliminating tertiary branches.
Fig 4B-E Indicate branch order in the images, this is unclear and a point that is focused on in the text. Done.
Discussion of Fig 1G from the text claims that shy87 is specifically required for branch elimination yet the data shows significant defects in branch outgrowth as well. This raises the question, are the branches abnormally stabilized that results in early underdevelopment and late atrophy? Authors should acknowledge alternative hypotheses. We agree and will revise the text accordingly. The difference between shy87 and control dauers, while statistically significant, is relatively minor and can only be detected by careful quantification, it is not apparent from looking at the images (in contrast for example to rab-8 and rab-10 mutants, where we acknowledge in the text that their branching defects might affect subsequent pruning.
Authors reference a branch elimination process but don't outline what this would entail and where their results fit in. We apologize for being unclear. Given that sax-1 and sax-2 function together, one would intuitively expect to see SAX-2 being reduced in sax-1 mutants, yet the opposite is observed. On potential explanation is that SAX-1 does not directly control SAX-2 abundance, but that clearance of SAX-2 is part of the pruning process that both proteins regulate. This would explain the enrichment of SAX-2 in sax-1 mutants. However, additional models cannot be excluded, and we acknowledge this in the revised text.
References:
Corchado, Johnny Cruz, Abhishiktha Godthi, Kavinila Selvarasu, and Veena Prahlad. 2024. “Robustness and Variability in Caenorhabditis Elegans Dauer Gene Expression.” Preprint, bioRxiv, August 26. https://doi.org/10.1101/2024.08.15.608164.
Karp, Xantha. 2018. “Working with Dauer Larvae.” WormBook, August 9, 1–19. https://doi.org/10.1895/wormbook.1.180.1.
Kozik, Patrycja, Richard W Francis, Matthew N J Seaman, and Margaret S Robinson. 2010. “A Screen for Endocytic Motifs.” Traffic (Copenhagen, Denmark) 11 (6): 843–55. https://doi.org/10.1111/j.1600-0854.2010.01056.x.
Lee, T., and L. Luo. 1999. “Mosaic Analysis with a Repressible Cell Marker for Studies of Gene Function in Neuronal Morphogenesis.” Neuron 22 (3): 451–61.
Swanson, M. M., and D. L. Riddle. 1981. “Critical Periods in the Development of the Caenorhabditis Elegans Dauer Larva.” Developmental Biology 84 (1): 27–40. https://doi.org/10.1016/0012-1606(81)90367-5.
Tang, Rui, Christopher W Murray, Ian L Linde, et al. n.d. “A Versatile System to Record Cell-Cell Interactions.” eLife 9: e61080. https://doi.org/10.7554/eLife.61080.
Zhao, Ting, Liying Guan, Xuehua Ma, Baohui Chen, Mei Ding, and Wei Zou. 2022. “The Cell Cortex-Localized Protein CHDP-1 Is Required for Dendritic Development and Transport in C. Elegans Neurons.” PLOS Genetics 18 (9): e1010381. https://doi.org/10.1371/journal.pgen.1010381.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
Figueroa-Delgado et al. use a C. elegans neuro plasticity model to examine how dendrites are eliminated upon recovery from the stress induced larval stage, dauer. The authors performed a mutagenesis screen to identify novel regulators of dendrite elimination and revealed some surprising results. Branch elimination mechanism varies between 2{degree sign}, 3{degree sign}, and 4{degree sign} branches. The NDR kinase, SAX-1 and it's interactors (SAX-2 and MOB-2) are required for elimination of second and third order branches but not fourth order branches. Interestingly they showed that branch elimination varies depending on the stimulus of dendrite outgrowth such that the NDR kinase is required for branch elimination after genetically inducing the dauer stage but is not required if dauers are produced through food deprivation. The authors go a step further to include a small candidate screen looking at various pathways of membrane remodeling and identify additional regulators of dendrite elimination related to membrane trafficking including RABI-1, RAB-8, RAB-10, and RAB-11.2.
Major comments:
Minor comments:
Referee cross-commenting
I agree that we shared many of the same concerns.
There are several general assays for dauer characteristics that could be used here to determine if the post-dauer animals retain other characteristics of the dauer stage in addition to IL2 branches (SDS resistance, alae remodeling, pharyngeal bulb morphology, nictation behavior). The nictation behavior has been connected very nicely with IL2 neurons (Junho Lee's group). Additionally, FLP dendrites occupy the same space as the IL2 branches and outgrowth in post-dauers occurs in coordination with IL2 branch elimination - this might be another optional experiment, to check if FLP growth is impeded by persistent IL2 branches. All of these could be quantified similar to how the authors have already established with their IL2 model (FLP dendrite branches) or with a binary statistic.
These results describe a new role for the NDR kinase complex in dendrite pruning that has clinical significance to our understanding of human brain development and human health concerns in which pruning is dysregulated, such as observed in the case of autism. The authors use an established neuro-plasticity, C. elegans model (Schroeder et al. 2013) which provides a tractable and reproduceable platform for discovering the mechanism of dendrite pruning. These results would influence future work in the fields of cell biology of the neuron and disease models of brain development.
My expertise is in the field of C. elegans neuroscience and stress biology and have sufficient expertise to evaluate all aspects of this work.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this manuscript, the authors establish C. elegans IL2 neurons as a system in which to study dendrite pruning. They use the system to perform a genetic screen for pruning regulators and find an allele of sax-1. Unexpectedly sax-1 is only required for post-dauer pruning in two different genetic backgrounds that induce dauer formation, but not starvation-induced dauer formation. Sax-1/NDR kinase reduction has previously been associated with increased outgrowth and branching in other systems, so this is a new role for this protein. However, the authors show that proteins that work with Sax-1 in other systems, like sax-2/fry, also play a role in this pathway. The genetic experiments are beautiful and the findings are all clearly explained and strongly supported. The authors also examine sax-2 localization, which localizes sax-1 in other systems, and show it in puncta in dendrites that increase with dauer exit, consistent with function at the time of pruning. They also show that membrane trafficking regulators associated with NDR kinases function in the same pathway here, hinting that endocytosis may play a role during pruning as in Drosophila. The link to endocytosis was a little weak (see Major point below). Overall, this study describes a new system to study pruning and identifies NDR/fry/Rabs as regulators of pruning during dauer exit. The work is very high quality and both the imaging and genetics are extremely well done.
Major points
Minor:
"shy87 mutant dauers showed a minor reduction in secondary and tertiary branches compared to control (Figure 1G). These results indicate that shy87 is specifically required for the elimination of dauer-generated dendrite branches." Maybe temper the specificity claim some as the reduction in branches is definitely there.
"three complimentary approaches" should be complementary
"In control animals, SAX-2 was mostly concentrated in the cell body (data not shown)" It might be nice to include some overview images that show the cell body for completeness.
Neurite pruning is important in all animals with neurons. Genetic approaches have primarily been applied to the problem using Drosophila, so identifying a new model system in which to study it is an important step. Using this system, a pathway known to function in a different context is linked to pruning. Thus the study provides new insights into both pruning and this pathway.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This interesting study uses an unbiased genetic screen in C. elegans to identify SAX-1/NDR kinase as a regulator of dendritic branch elimination. Loss of SAX-1 results in an excess branching phenotype that is striking and highly penetrant. The authors identify several additional regulators of branch elimination (SAX-2, MOB-1, RABI-1, RAB-11.2) by using a candidate genetic screen aimed at factors that interact physically or genetically with SAX-1. They propose that SAX-1 acts by promoting membrane retrieval based on the nature of these interactors and the results of an imaging-based in vivo assay for endocytic puncta.
Major comments.
While this does not undermine the importance of the results, it does require more explanation. The authors write that "the requirement for sax-1... relies on specific physiological states of the dauer stage," but I do not understand what this means. Are they saying that daf-7 and daf-2 dauers are in a different "physiological state" than wild-type dauers? In what way? What is the evidence for this? A more rigorous explanation is needed.
To me, the simplest genetic explanation is that daf-7 and daf-2 are partially required for branch retraction in a manner redundant with sax-1, and the ts mutants are not fully wild-type at 15C. Thus, the sax-1 requirement is revealed only in these mutant backgrounds. Can the authors examine starvation-induced dauers of daf-7 or daf-2 raised continuously at 15C?
daf-7 and daf-2 ts strains can form "partial dauers" that have a dauer-like appearance but are not SDS resistant. Could the difference between partial dauers and full dauers account for the difference in sax-1-dependence? The authors could use SDS selection of the daf-7 strain at 25C to ensure they are examining full dauers.
The Bargmann lab has created a daf-2 FLP-OUT strain (ky1095ky1087) that allows cell-type-specific removal of daf-2. Could this be used to test for a cell-autonomous role of daf-2 in IL2Q related to branch elimination?
These ideas are not a list of specific experiments the authors need to complete, rather they are meant to illustrate some possible approaches to the question. Whatever approach they use, it is important for them to more rigorously explain why SAX-1 is not required for branch removal in wild-type animals. 2. The SAX-2 localization (Fig. 4) and endocytosis assay (Fig. 6) results were not clear to me from the data shown. Overall a more rigorous analysis and presentation of the data would be important to make these conclusions convincing. This may involve refining the data presentation in the figures, modifying the claims (e.g., "we propose" vs "we find"), or saving some of the data to be more fully explored in a future paper. In my view, these figures are the biggest weak point of the manuscript and also are not important for the central conclusions (which are well supported and convincing), indeed these results are barely mentioned in the Abstract or last paragraph of Introduction.
Minor points:
Referee cross-commenting
I think we all touched on similar points. I wanted to follow up on Reviewer 3's comment, "Is the failure to eliminate branches an indication of incomplete dauer recovery? Do sax-1 mutants retain additional characteristics of dauer morphology in post dauer adults." I thought this was an excellent point. It made me wonder if that might explain why the defect is only seen in daf-7 and daf-2 mutant backgrounds - maybe these strains retain partial dauer traits even after exit. Is there a specific experiment that they could do? Did you have specific characteristics of dauer morphology in mind for them to check? (Ideally something in the nervous system that can be scored quantitatively.)
A major strength of this work is the pioneering use of a novel system to study neuronal branch retraction. C. elegans has provided a powerful model for studying how dendrite branches form, but much less attention has been paid to how excess neuronal branches are removed. The post-dauer remodeling of IL2Q neurons provides an exciting and dramatic physiological example to explore this question.
This paper is notable for taking the first steps towards developing this innovative model. It does exactly what is needed at the outset of a new exploration - a forward genetic screen to discover the main regulators of the process. Using a combination of classical and modern genetic approaches, the authors bootstrap their way to a sizeable list of factors and a solid understanding of the properties of this system, for example that retraction of higher vs lower order dendrites show different genetic requirements.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
We thank the reviewers for their positive comments. Our manuscript is to our knowledge the first to investigate the role of VAIL (V-ATPase—ATG16L1 induced LC3 lipidation), a form of CASM (Conjugation of ATG8s to single membranes) in SARS-CoV-2 replication. We demonstrate that SARS-CoV-2 Envelope (E) induces VAIL and this contributes to viral replication, including by using a reverse genetics system to make an E mutant virus. There have been many high quality studies examining the role of canonical autophagy in SARS-CoV-2 replication and our manuscript does not argue that all or even most LC3 lipidation during infection is via VAIL. We will try to make this point more clearly in the text. We do not think this detracts from the novelty and importance of our manuscript.
*Reviewer #1 (Evidence, reproducibility and clarity (Required)): *
Figueras-Novoa et al present a short report demonstrating the induction of LC3 lipidation on single membranes by SARS-CoV-2 through a noncanonical autophagy pathway referred to as VAIL. The authors utilize elegant genetic tools to show that the induction of LC3 lipidation upon viral infection is mainly due to VAIL rather than canonical autophagy. They demonstrate that the activity of the viral E protein that can cause neutralization of acidic vesicles leads to the activation of non-canonical LC3 lipidation on single membranes. Interestingly, the authors also conclude that the impairment of VAIL leads to a reduction of viral load as a result of a defect in later stages of viral infection, although the underlying mechanism was not further explored. *
Overall, this is an elegant and well controlled study that provides a clear conclusion. I only have some minor comments.*
We thank the reviewer for their assessment of our manuscript.
In some experiments, LC3 lipidation does not appear to be fully disrupted upon VAIL inhibition (e.g. Fig.'s 1H, 3D, 4A). As other labs have shown that SARS-CoV2 blocks autophagic flux, this could be further clarified in this manuscript as both VAIL and autophagy may be co-induced upon viral infection.
We agree with the reviewer that there is a contribution of canonical macroautophagy to the LC3B lipidation observed in SARS-CoV-2. We will extend the discussion in the manuscript to clarify this point for the readers.
Can the authors test the induction of LC3 lipidation in cells expressing K490 mutant of ATG16L1 in ATG16L1 KO cells to compare them with ATG16L1-ATG13 double knockouts?
The western blot in figure 3F (quantified in Figure 3G) shows LC3B lipidation in response to E expression in ATG16L1-ATG13 double knock out cells reconstituted with wild type ATG16L1 but not in cells complimented with ATG16L1 K490A mutant. We agree that the referee’s suggestion to perform these experiments in the context of infection would be informative. However in spite of numerous attempts, we have so far been unable to generate a cell clone fully devoid of ATG16L1 in a cell line that can be productively infected with SARS-CoV-2. For reasons unclear to us there appears to be a very low level of residual ATG16L1 activity despite multiple different CRISPR/Cas9 targeting attempts. The suggested complementation experiments might still be informative in the context of low level ATG16L1 expression so we will pursue this. Alternatively, as a contingency we can try to produce SARS-CoV-2 infectable cells with mutations in ATG16L1’s binding partner V1H, this interaction is required for VAIL. A further contingency could be to assess LC3B lipidation during infection and treatment with a Vps34 inhibitor, which inhibits canonical autophagy.
Minor points: * * The difference between Fig. 1F&G is unclear and why the authors are including both analyses. Similarly figures 4G&H.
We included both metrics to show that the decrease in LC3B lipidation in cells expressing SopF during infection is robust and observed in two separate readouts. While spot area measures the area of infected cells covered by GFP-LC3B fluorescence, spot intensity is a reading of the intensity of the area defined in an infected cell as being LC3 positive. Theoretically, these measurements could change in different ways. For example, if the same amount of lipidated LC3 were to distribute over a larger area of the cell. We prefer to keep both measurements in the manuscript.
The authors should show boxed colocalisation of all images, including negative controls. For examples, the authors have shown boxed magnifications in only the lowest panel in Figure 2A but not the upper two panels. Figures 4E&F should include boxed examples. This serves to clarify both positive and negative colocalisation events.
Boxed magnifications will be added to all images.
Reviewer #1 (Significance (Required)): *
Overall an elegant and well controlled study demonstrating the induction of non-canonical LC3 conjugation on single membranes (VAIL) during SARS-CoV2 infection. A further exploration of canonical autophagy (as previously published by others) in addition to VAIL would enhance this study.*
As the reviewer noted, several excellent studies have explored canonical autophagy during SARS-CoV-2 infection, many of which we cite in our manuscript. Our focus, however, is to demonstrate that SARS-CoV-2 E induces LC3 lipidation via VAIL. We believe that exploring the diverse roles of canonical autophagy mechanisms in SARS-CoV-2 infection is beyond the scope of this study.
*This study is of interest to researchers studying autophagy, viruses, immunology, single membrane LC3 lipidation, and lysosomes as well as potentially clinicians treating SARS-CoV2 infecteted individuals. *
We thank the reviewer for this assessment of our manuscript.
*Reviewer #2 (Evidence, reproducibility and clarity (Required)): *
Major Comments *
Figure 1D does not very clearly show an overlap between V1D and LC3B. Both proteins seem broadly present across the cell and there is no easily identifiable change in V1D distribution upon infection. As such the overlay may be purely stochastic. The authors should quantify the observed co-localization events across multiple cells and biological replicates and compare them to other protein(s) with a similar cellular distribution pattern.*
We agree there is no obvious change in V1D staining on infection. The images in Figure 1D are purely intended to illustrate that LC3 and the V-ATPase can colocalise, not to demonstrate a change in V-ATPase distribution or to suggest a direct interaction. We will make this point more clearly in the text. We will also carry out analyses of the kind (see also response to the first two Minor Comments). We would be happy to provide an alternative method of visualising the V-ATPase (we could use any suitable antibody to the V-ATPase, or the bacterial effector SidK) if required. In response to reviewer 3’s comments, we will carry out a pull-down experiment to test the association of the V-ATPase and ATG16L1 during E expression, as this is a key interaction during VAIL activation.
Based on Figure 2F the authors suggest that virus entry is unaffected by the inhibition of VAIL in early timepoints. However, according to the figure legend, the timepoint used is 7hpi, while 2D uses 24hpi. Some SARS-CoV-2 papers suggest 7-10 hours is sufficient time to release new virions (Ban-On et al., 2020). As such 7hpi can not necessarily be seen as an early time point. Did the authors test earlier ones? Also, based on this, would it be possible that the effects observed at 24hpi are actually secondary infections, meaning that the virus utilizes pathway components for virion production and a lack thereof reduces infectivity of newly formed virions? In this case it would be interesting to set up an assay that can distinguish between primary and secondary infection to study both individually more closely.
Whereas 7 hours may be sufficient to release new virions, it is not sufficient to establish infections in other cells – this is why we chose that time point. The observation that there is no difference in the percentage of infected cells at 7 h p.i. (figure 2F) led us to suggest that viral entry is unaffected . We then confirmed this through the pseudovirus assay in Figure 2G, where no difference is found between SopF and mCherry expressing cells. For this assay, GFP-expressing, replication incompetent, lentiviral particles pseudotyped with Spike from different SARS-CoV-2 lineages were used to transduce mCherry and SopF expressing cells. A change in the percentage of GFP-positive cells would indicate an effect on viral entry, but no such change was observed in SopF-expressing cells.
We agree with the reviewer that the effects observed at 24 hpi are likely due to a defect in subsequent rounds of infection, since no difference was observed at 7 hpi or with our pseudovirus assay. We will attempt to make this point in the text as clearly as possible.
The authors nicely show in their study an involvement of VAIL in SARS-CoV-2 mediated LC3 lipidation. However, the observed effects are relatively moderate in several experiments, indicating that there may be another contributor to the observed phenotype. It would be nice to highlight this in the discussion and debate potential mechanisms that are causing the observed effects during infection.
We agree with the reviewer’s analysis. We have discussed the contribution of canonical autophagy in the second paragraph of the discussion, but we will expand on this in a revised manuscript. E expression levels are moderate during infection, other structural proteins such as N and M are present in much higher amounts. Since E is the key protein in VAIL initiation, a moderate effect of VAIL inhibition in perhaps expected. Nonetheless this still plays a crucial role in the viral life cycle.
*Minor Comments *
This quantification of GFP-LC3 relocalisation will be carried out and included.
The quantification of V1D, E and GFP-LC3 will be carried out and included.
For Figure 2H-K the authors perform KDs of ATG16L1 and ATG13. While the results for the two specific proteins are certainly convincing, the authors would strengthen their argument by testing additional proteins in the autophagy pathway to support their claim that VAIL but not autophagy affects protein abundance of N (OPTIONAL).
As discussed in response to reviewer 1, we will attempt to infect ATG16L1 KO cells reconstituted with a K490A ATG16L1 mutant, which is an established tool and has been validated to be deficient in VAIL but not canonical autophagy.
***Referee cross-commenting** *
As outlined above in response to reviewer 1 and below to reviewer 3, we agree that there is a modest contribution of VAIL to overall LC3 lipidation, which correlates with a modest amount of E expression in SARS-CoV-2 infection. VAIL is clearly important for the viral life cycle, thus whatever the proportion of LC3 lipidation attributable to this pathway it must be biologically significant.
*Reviewer #2 (Significance (Required)): *
While previous publications have shown interaction between SARS-CoV2 and autophagy, the authors of this manuscript demonstrate that V-ATPase-ATG16L1 induced LC3 lipidation (VAIL) is activated during infection and affects viral replication. *
This study provides an interesting new aspect to host-SARS_CoV-2 interactions. *
The manuscript is of interest for people studying virus-host cell interaction, as well as for researchers in the fields of infectious diseases, specifically SARS-CoV2, and autophagy/VAIL*.
We thank the reviewer for their assessment of our manuscript.
R*eviewer #3 (Evidence, reproducibility and clarity (Required)): *
The interaction of SARS-CoV-2 with canonical autophagy has been well documented. However, whether SARS-CoV-2 infection induces and benefits from non-canonical autophagy is unclear. In this manuscript, the authors demonstrated that SARS-CoV-2 infection induces V-ATPase-ATG16L1-induced LC3 lipidation (VAIL), a form of non-canonical autophagy in which LC3 is conjugated to single membranes. The SARS-CoV-2 envelope protein, through its ion channel activity, triggers the V-ATPase proton pump and induces VAIL during SARS-CoV-2 infection. Inhibiting VAIL during SARS-CoV-2 infection with SopF, a Salmonella effector, attenuates SARS-CoV-2 egress. *
While these findings are interesting and demonstrate that SARS-CoV-2 infection triggers VAIL for its own benefit, the mechanism by which VAIL promotes SARS-CoV-2 replication remains unclear. Moreover, the contribution of VAIL to LC3 lipidation during SARS-CoV-2 infection appears to be minimal, as blocking VAIL through SoPF expression only marginally reduced LC3B lipidation (Fig. 1H). Therefore, the contribution of VAIL to LC3 lipidation during SARS-CoV-2 infection is minimal.*
We thank the reviewer for their assessment of our manuscript. As we have already alluded to in our response, we agree that only part of the LC3 lipidation observed during infection can be attributed to VAIL. There is a reproducible effect on viral replication which we have demonstrated in multiple ways, therefore the contribution of VAIL is of biological importance.
*Comments: *
We agree with the reviewer that this would be an informative experiment. We can carry out this experiment in an E expression system, rather than infection. This is due to the difficulty of getting enough material to carry out this kind of pull-down experiment in infected cells (at the time of writing these experiments still have to be carried out in CL3).
This is a reflection of multi-cycle kinetics. N is still very strongly expressed in infected cells, even after virions have egressed. SARS-CoV-2 can infect VAIL-deficient cells and expresses the same levels of N prior to subsequent rounds of infection (at 7 hours after infection for example). Egress in VAIL-deficient, SopF-expressing cells is defective. Therefore, fewer cells will be infected in subsequent rounds of infection in SopF expressing cells, resulting in fewer N-positive cells in the SopF expressing cell population (most obvious after 24 hours).
Figure 2H. The authors show that knockdown of ATG16L1 reduces the expression of N during SARS-CoV-2 infection compared to the controls. To confirm that knockdown of ATG16L1, which is required for both canonical autophagy and VAIL, reduces N staining via VAIL, the authors should examine the impact of SopF expression on N levels in ATG16L KD cells. This experiment will confirm if the reduction in N staining in ATG16L1 KD cells is due to VAIL.
As stated in the response to reviewer 1, we can attempt this experiment in an ATG16L1 KO system complemented with K490A ATG16L1, which is deficient in VAIL and not canonical autophagy.
In this western the exposure is deliberately turned up to show that minimal ATG13 was left after knock down. We will also show the full blot with less exposure – this will demonstrate high quality.
Also, N appears as a single band in Figure 2J, but appears as double bands in Figures 2A and H. Could the authors explain this?
An extra band can be seen in 2J for N. However, as the reviewer points out, the intensity of the lower band is fainter than in 2A or 2H. The biology of SARS-CoV-2 N is interesting and complicated, with different truncated isoforms and phosphorylation patterns observed (see for example Mears et al., 2025 PMID:39836705). We observed changes in abundance of the second band between experiments, but this did not obviously depend on VAIL. We therefore consider this to be beyond the scope of this investigation.
*Reviewer #3 (Significance (Required)): *
Our experiments show unambiguously that VAIL contributes to viral replication. Therefore even if As alluded to above, we do not think a further investigation of canonical macroautophagy and SARS-CoV-2 would enhance the quality of our manuscript. We will try to make our description of the contribution of macroautophagy clearer in the revised manuscript (without providing a full literature review). We also do not think that exploring the nature of the multiple N bands on western blot is within the scope of this paper.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The interaction of SARS-CoV-2 with canonical autophagy has been well documented. However, whether SARS-CoV-2 infection induces and benefits from non-canonical autophagy is unclear. In this manuscript, the authors demonstrated that SARS-CoV-2 infection induces V-ATPase-ATG16L1-induced LC3 lipidation (VAIL), a form of non-canonical autophagy in which LC3 is conjugated to single membranes. The SARS-CoV-2 envelope protein, through its ion channel activity, triggers the V-ATPase proton pump and induces VAIL during SARS-CoV-2 infection. Inhibiting VAIL during SARS-CoV-2 infection with SopF, a Salmonella effector, attenuates SARS-CoV-2 egress.
While these findings are interesting and demonstrate that SARS-CoV-2 infection triggers VAIL for its own benefit, the mechanism by which VAIL promotes SARS-CoV-2 replication remains unclear. Moreover, the contribution of VAIL to LC3 lipidation during SARS-CoV-2 infection appears to be minimal, as blocking VAIL through SoPF expression only marginally reduced LC3B lipidation (Fig. 1H). Therefore, the contribution of VAIL to LC3 lipidation during SARS-CoV-2 infection is minimal.
Comments:
The authors show that the ion channel activity of E is essential for VAIL induction during SARS-CoV-2 infection. Since V-ATPase recruits the ATG16L complex to induce VAIL, and to clarify how SARS-CoV-2 infection triggers VAIL, the authors should examine whether SARS-CoV-2 infection or the expression of E induces V-ATPase-ATG16L interaction and whether this interaction is disrupted when SopF is expressed.
Since the authors suggest that expression of SopF attenuates viral exit, one would expect that the number of N-positive cells will increase in SopF-expressing cells compared to the mCherry control cells. However, as shown in Figure 2D, this is not the case. Could the authors discuss why N-positive cells will be reduced in SopF-expressing cells when viral egress is impeded in these cells?
Figure 2H. The authors show that knockdown of ATG16L1 reduces the expression of N during SARS-CoV-2 infection compared to the controls. To confirm that knockdown of ATG16L1, which is required for both canonical autophagy and VAIL, reduces N staining via VAIL, the authors should examine the impact of SopF expression on N levels in ATG16L KD cells. This experiment will confirm if the reduction in N staining in ATG16L1 KD cells is due to VAIL.
Figure 2J. The quality of the Western blot data is poor. Also, N appears as a single band in Figure 2J, but appears as double bands in Figures 2A and H. Could the authors explain this?
This manuscript proposes a role for VAIL in LC3 lipidation during SARS-CoV-2 infection. While the findings are interesting, VAIL only marginally contributes to LC3 lipidation during SARS-CoV-2 infection. Therefore, the significance of VAIL to LC3B lipidation during SARS-CoV-2 infection is unclear.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Major Comments
Figure 1D does not very clearly show an overlap between V1D and LC3B. Both proteins seem broadly present across the cell and there is no easily identifiable change in V1D distribution upon infection. As such the overlay may be purely stochastic. The authors should quantify the observed co-localization events across multiple cells and biological replicates and compare them to other protein(s) with a similar cellular distribution pattern.
Based on Figure 2F the authors suggest that virus entry is unaffected by the inhibition of VAIL in early timepoints. However, according to the figure legend, the timepoint used is 7hpi, while 2D uses 24hpi. Some SARS-CoV-2 papers suggest 7-10 hours is sufficient time to release new virions (Ban-On et al., 2020). As such 7hpi can not necessarily be seen as an early time point. Did the authors test earlier ones? Also, based on this, would it be possible that the effects observed at 24hpi are actually secondary infections, meaning that the virus utilizes pathway components for virion production and a lack thereof reduces infectivity of newly formed virions? In this case it would be interesting to set up an assay that can distinguish between primary and secondary infection to study both individually more closely.
The authors nicely show in their study an involvement of VAIL in SARS-CoV-2 mediated LC3 lipidation. However, the observed effects are relatively moderate in several experiments, indicating that there may be another contributor to the observed phenotype. It would be nice to highlight this in the discussion and debate potential mechanisms that are causing the observed effects during infection.
Minor Comments
The re-localization events shown in Fig 3A should be quantified.
The co-localization events displayed in Fig 4A should be quantified.
For Figure 2H-K the authors perform KDs of ATG16L1 and ATG13. While the results for the two specific proteins are certainly convincing, the authors would strengthen their argument by testing additional proteins in the autophagy pathway to support their claim that VAIL but not autophagy affects protein abundance of N (OPTIONAL).
Referee cross-commenting
Overall I agree with the comments of my co-reviewers and I think the suggested experiments/comments are sensible. I in part already eluted to it my analysis, but I tend to agree with reviewer 3 on the limited effect VAIL seems to have on LC3b lipidation.
While previous publications have shown interaction between SARS-CoV2 and autophagy, the authors of this manuscript demonstrate that V-ATPase-ATG16L1 induced LC3 lipidation (VAIL) is activated during infection and affects viral replication.
This study provides an interesting new aspect to host-SARS_CoV-2 interactions.
The manuscript is of interest for people studying virus-host cell interaction, as well as for researchers in the fields of infectious diseases, specifically SARS-CoV2, and autophagy/VAIL.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Figueras-Novoa et al present a short report demonstrating the induction of LC3 lipidation on single membranes by SARS-CoV-2 through a noncanonical autophagy pathway referred to as VAIL. The authors utilize elegant genetic tools to show that the induction of LC3 lipidation upon viral infection is mainly due to VAIL rather than canonical autophagy. They demonstrate that the activity of the viral E protein that can cause neutralization of acidic vesicles leads to the activation of non-canonical LC3 lipidation on single membranes. Interestingly, the authors also conclude that the impairment of VAIL leads to a reduction of viral load as a result of a defect in later stages of viral infection, although the underlying mechanism was not further explored.
Overall, this is an elegant and well controlled study that provides a clear conclusion. I only have some minor comments.
In some experiments, LC3 lipidation does not appear to be fully disrupted upon VAIL inhibition (e.g. Fig.'s 1H, 3D, 4A). As other labs have shown that SARS-CoV2 blocks autophagic flux, this could be further clarified in this manuscript as both VAIL and autophagy may be co-induced upon viral infection. Can the authors test the induction of LC3 lipidation in cells expressing K490 mutant of ATG16L1 in ATG16L1 KO cells to compare them with ATG16L1-ATG13 double knockouts?
Minor points:
The difference between Fig. 1F&G is unclear and why the authors are including both analyses. Similarly figures 4G&H.
The authors should show boxed colocalisation of all images, including negative controls. For examples, the authors have shown boxed magnifications in only the lowest panel in Figure 2A but not the upper two panels. Figures 4E&F should include boxed examples. This serves to clarify both positive and negative colocalisation events.
Overall an elegant and well controlled study demonstrating the induction of non-canonical LC3 conjugation on single membranes (VAIL) during SARS-CoV2 infection. A further exploration of canonical autophagy (as previously published by others) in addition to VAIL would enhance this study.
This study is of interest to researchers studying autophagy, viruses, immunology, single membrane LC3 lipidation, and lysosomes as well as potentially clinicians treating SARS-CoV2 infecteted individuals.
This reviewer is experienced in autophagy research.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
We appreciate the constructive and supportive feedback on our manuscript. All three reviewers acknowledged the significance and novelty of our work on bacterial telomere protection. In response to their suggestions, we have conducted the requested experiments and revised the manuscript accordingly. These changes have enhanced the rigor of our study and clarified our interpretations and explanations.
Moreover, we characterized an additional truncation mutant of TelN (TelN Δ445–631), which lacks the two C-terminal domains. Despite this deletion, the mutant retained protection activity (Supplementary Figure S4B), indicating that the remaining regions of the protein are sufficient to confer efficient protection in this assay.
Finally, we removed three sequence alignments (previously Supplementary Figures S6A and S7), as we recognized that the high degree of sequence divergence could hinder proper alignment and potentially lead to misinterpretation.
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
This study addresses how the bacterial telomere protein TelN protects telomere ends against the action of the Mre11-Rad50 nuclease (MR). This protection is essential for the stability of hairpin-ended linear plasmid and chromosomes in bacteria but had not been explored before. The authors demonstrate that TelN is necessary and sufficient to block MR-dependent DNA cleavage when bound to its specific telomere sequence. By combining elegant genetics and biochemical approaches, it convincingly shows that TelN-dependent inhibition likely involves a specific interaction between TelN and the MR complex. The manuscript is well written, easy to read and focused on the relevant information. The claims and the conclusions are supported by the data. There is no over-interpretation.
Comments: - Figure 1B, unnormalized transformation efficiency would be useful to show in SI
The unnormalized B. subtilis transformation efficiency has now been added as new figure panel S1B.
- Figures 2B, 2C, 3C, 3D, 4C, 5A and 5B: quantification of independent experiments should be added
While these DNA protection experiments show a clearly reproducible pattern of DNA degradation, the exact response to TelN titration varies somewhat between experimental replicates. We initially included the quantification of remaining full-length DNA because the corresponding band is hard to discern in the gel image due to pixel saturation. However, we realize now that this may mislead readers to think that the degradation occurs always with the exact same dosage response.
To avoid this, we have decided to remove the quantification and instead show the relevant part of the gel also at higher contrast to better visualize the loss of full-length DNA due to DNA degradation. In addition, we have included replicate experiments carried out at the same MR concentration (125 nM M₂R₂) or at higher concentration (500 nM M₂R₂) in the supplementary material. These examples demonstrate the general reproducibility of the assay.
**Referee cross-commenting**
Perfect for me. It seems that there is a consensus.
Reviewer #1 (Significance (Required)):
This pioneering study provides a very strong basis for a new understanding of telomeres in bacteria and offers fascinating evolutionary perspectives when compared to similar mechanisms active at telomeres in eukaryotic cells.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
The paper is well-presented and well-written throughout. The paper shows convincingly that TelN protects hairpin DNA ends from the activity of SbcCD, presumably providing a protection mechanism for N15 phage DNA in vivo. Furthermore, this protection activity is shown not to require the catalytic (resolvase) activity of TelN, nor its poorly characterised C-terminal domain. The paper also suggests that this inhibition acts both at the level of competition for the DNA hairpin end and at the level of a direct protein:protein interaction between TelN and MR. An (acknowledged) weakness is that there is no real insight into the protein:protein interaction suggested by the experiments shown in Figure 5. Ideally, the protein:protein interaction interface would be identified and mutations in this interface would be shown to reduce hairpin protection.
Specific comments/questions
(1) What pathway (in vivo) leads to inactivation of linear hairpin DNA - one suspects that cleavage by SbcCD at the hairpins is probably not the full story. Presumably SbcCD cleavage facilitates further processing by other long range resection systems such as RecBCD, Exo1, RecQ/J etc. Would it be appropriate to view the hairpin as an adaption to protect against these nucleases, which then must be complemented with a mechanism to suppress SbcCD?
The reviewer's suggestion that hairpin ends represent a first layer of adaptation against nucleolytic processing is compelling. Hairpin structures inherently resist many exonucleases due to their covalently closed nature (absence of free 3’ or 5’ ends) but remain vulnerable to MR processing (Connelly et al, 1998, 1999; Saathoff et al, 2018). This creates a scenario where effective telomere protection requires both the structural barrier provided by the hairpin and an active mechanism to suppress MR activity. We have added this perspective to the relevant paragraph in the discussion.
(2) Section starting "Direct inhibition of MR by TelN in vitro". What is the word direct supposed to convey here? To me it suggests that the inhibition is via direct interaction of TelN with MR (rather than, for example, a result of competition for the hairpin DNA end) which is not shown here. Suggest either defining or removing the word direct. This point gains more importance considering that differentiating between inhibition mechanisms becomes a focus of later parts of the paper.
By "direct inhibition," we meant that TelN blocks MR nuclease activity without requiring additional cofactors, as demonstrated in this minimal reaction system containing only TelN, MR complex, DNA substrate, and ATP. To avoid ambiguity, we have reworded the corresponding headline and paragraph.
(3) Figure 2B - Why no control lane without MR? - this is a basic control to show that he degradation we are seeing in the absence of TelN is MR-dependent. Formally, as shown, the degradation could be caused by the ATP stock.
We have now included ATP-only control lanes (without MR complex), which show no substrate degradation, confirming that ATP stocks do not contain contaminating nucleases and that the observed degradation is indeed MR-dependent. These controls are included in the supplementary data (Figure S3A) along with additional replicate experiments. Notably, the dose-dependent protection observed at low TelN concentrations (where MR activity is not fully inhibited) provides additional evidence for the specificity of the MR-TelN interaction system, as non-specific nuclease contamination would result in complete substrate degradation regardless of TelN concentration.
(4) Why not use B. subtilis SbcCD for the species specificity experiment? Also, is it not surprising that TelN yielded zero protection against MRX given that the DNA sequence specificity experiments above suggest competition for DNA substrate is part of the inhibition mechanism?
We agree that this would be a great addition. We attempted but were unable to purify active B. subtilis SbcCD protein despite multiple attempts. The yeast MRX experiment serves the same purpose of demonstrating species specificity and represents a more evolutionarily distant comparison, which strengthens our conclusions about bacterial-specific inhibition.
(5) If the authors felt it appropriate, I thought there was scope for further discussion/introductory material. There are strong parallels here with mechanisms used by phage to protect themselves from the activities of RecBCD, which include both proteins that protect DNA ends like T4 gene 2, we well as proteins that bind directly to RecBCD to inactivate it like lambda Gam. As such, the work here will appeal as much to those interested in bacterial defence systems / phage:host interactions as it does to those interested in telomere biology. Especially significant is the inhibition of DNA end processing factors by lambda Gam since this protein is reported to interact with both RecBCD and SbcCD (PMID: 2531105).
We agree that there are obvious parallels between lambda Gam and TelN as counter-defence factors. This was likely largely missed in previous work because the telomere resolution activity of TelN masked its function in counter-defence. We have added a statement on this matter at the end of the discussion.
(6) Just a gripe really: it seems to be 'de rigeur' at the moment to re-name bacterial proteins for their human orthologues, presumably to elevate the perceived importance of the work(?), but it is not a practice I think is terribly helpful as it causes issues when searching literature. Minimally it would be great if the authors could ensure they add SbcCD as a keyword for search purposes.
We appreciate the reviewer's concern about nomenclature inconsistencies in the literature. We have chosen MR over SbcCD as a more generic term that covers eukaryotes, archaea and lately also bacteria and will hopefully contribute to a more consistent terminology in the literature across the domains of life in the future. Our choice to use "Mre11-Rad50" (MR) for the E. coli SbcCD complex is also consistent with prominent recent publications (Käshammer et al., 2019; Gut et al., 2022), explicitly referring to the E. coli system as "Mre11-Rad50" while acknowledging the bacterial designation. To link to previous literature, we made sure that both "SbcCD" and "Mre11-Rad50" are mentioned in the abstract. And, as suggested, we have now also added “SbcCD” to our keyword list to facilitate comprehensive literature searches.
**Referee cross-commenting**
I have nothing to add. The reviewers' comments are all broadly positive and consistent.
Reviewer #2 (Significance (Required):
This is an excellent paper unveiling a phage encoded "counter-defence" mechanism designed to protect phage DNA from degradation. It will be of special interest to those studying telomere biology of phage:host interactions.
Reviewer #3
The authors investigate how the N15 phage protelomerase TelN protects linear chromosomes that terminate in hairpin structures (a sort of telomere). In E. coli and B. subtilis cells, removal or truncation of telN reduces transformation/survival of linear DNA, whereas complementation with full-length or a catalytically inactive TelN restores viability, consistent with TelN playing a nonenzymatic capping function.
In vitro, TelN binds hairpin substrates with moderate affinity and protects them from the nuclease activity of the Mre11/Rad50 complex. The authors propose that TelN originated as an early, sequence specific barrier against MR mediated DNA end processing, establishing fundamental principles of telomere protection that persist from bacteria to eukaryotes.
Major comments:
The manuscript convincingly shows that TelN can functionally block the Mre11Rad50 (MR) nuclease on a hairpin DNA end in a sequence specific manner (suggesting a physical interaction), but it doesn't directly demonstrate this. A simple pull-down or equilibrium binding method would be useful in proving a physical interaction.
We agree that this would be a valuable addition to the study. We have made several attempts to detect direct interaction by co-immunoprecipitation. However, without success so far. We do not have sufficient material for equilibrium binding methods (yet).__ ____ __
The MR complex requires ATP hydrolysis for resection of DNA ends. It would be a nice addition to the manuscript if the effect of TelN of Rad50 ATPase activity was tested.
We have tested the effect of TelN on Rad50 ATPase activity and found no significant impact under our experimental conditions, possible in line with the lack of stable interaction.
The bar plot on Fig 3B indicates that the experiments are performed in triplicate. The statistical significance of the differences between conditions should be determined. The same general comment could be made regarding the quantification of the polyacrylamide gels - how reproducible are these values?
We performed paired t-test analysis for the following figures and now indicate the p-values wherever significant (below 0.05): Figures 1D, 1E, 3B, 4B and S4B. We used paired t-tests to generally compare linear vs circular plasmid transformation efficiency for each condition. In Figure 4B, which included two different linear DNA constructs, we compared the two linear DNA constructs directly to each other. [Given that our experimental design included multiple control conditions with known expected outcomes to validate assay performance, rather than many independent exploratory comparisons, we report uncorrected p-values as the primary analysis. The inclusion of multiple controls with predictable outcomes reduces the likelihood of false positive interpretations.]
As stated in response to reviewer 1, while the exact values for the DNA degradation profile vary somewhat between experiments (likely due to variations in band quantification – see also response to comment below), the general trends are robust as for example indicated by similar experiments performed with higher MR concentration (500 nM instead of 125 nM M₂R₂ concentrations for all TelN variants) demonstrating reproducibility across different conditions. For Figure 5, however, we are unable to provide additional repeat experiments due to limitations in reagent availability. Considering the robust effect seen with Ec MR controls and the presence of multiple samples in the dilution series, we are nevertheless confident about the conclusion.
Minor comments:
A better explanation of how the gels were quantified should be provided. Were the products included in the analysis, or was it just the decrease in the substrate band that was measured?
As also stated above, we have removed the band quantification and instead show the bands also at different contrast settings.
In our original approach, gel band quantification was performed using ImageQuant TL software (version 8.2.0, GE Healthcare). For each gel, individual lanes were defined using either fixed-width boundaries (95-103 pixels) or automatic edge detection, depending on the gel quality and band definition. Band volumes were calculated using rolling ball background subtraction (radius 180 pixels) with automatic band detection. Substrate degradation was assessed by measuring the integrated density (volume) of the remaining full-length (or near full-length) substrate bands under different treatment conditions. The band volume values were plotted directly to compare substrate levels across treatment groups.
We now present the data as two gel panels: an exposure showing the full reaction profile, and another exposure focusing on the substrate bands to clearly demonstrate dose-dependent protection. Additional replicate experiments including ATP-only controls (confirming no contamination from ATP stocks) and experiments at 500 nM M₂R₂ concentrations, are provided in the supplementary data. This approach provides more direct visualization of the biological phenomenon with comprehensive control validation.
I felt like the Results jump rather abruptly from B. subtilis chromosome assays to E. coli plasmid experiments. Maybe the addition of a few linking sentences would improve this transition.
Upon re-reading the manuscript we agree with this assertion and have added further information to provide a smoother transition.
A comment on the stoichiometry of TelN and genome ends during phage replication would be useful.
Our in vitro data suggest that effective protection can be achieved at relatively low TelN:DNA ratios in vitro, consistent with the notion of formation of stable, protective nucleoprotein structures. We unfortunately do not currently have information on the copy number of TelN per cell or per hairpin end. It is not easy to obtain reliable values for these numbers. However, we can speculate that multiple TelN proteins are present due to the presence of three copies of a DNA sequence motif (binding to CTD1) in each telomeric DNA, consistent with the formation of stable, protective nucleoprotein structures.
Reviewer #3 (Significance (Required)):
General assessment:
Strengths: A nice combination of genetics and biochemistry convincingly demonstrates that TelN protects linear chromosomes/replicons from MR-dependent degradation independent of its cleavage-ligase activity. It does this by binding to the hairpin DNA ends in a sequence specific fashion and the species specificity suggests a direct physical interaction, which likely inhibits the nuclease activity of the MR complex
Limitations: The lack of characterization of the putative physical interaction between TelN and the MR complex is considered a weakness.
Advance: The manuscript fills in a mechanistic gap between protelomerase-mediated telomere formation and maintenance by demonstrating a protective/capping role. This is the first quantitative analysis of DNA-end protection from MR nuclease activity by TelN.
Audience: Readers interested in bacterial chromosome biology, DNA repair, the parallels to eukaryotic shelterin will be interesting to the broader telomere and genome stability communities.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
The authors investigate how the N15 phage protelomerase TelN protects linear chromosomes that terminate in hairpin structures (a sort of telomere). In E. coli and B. subtilis cells, removal or truncation of telN reduces transformation/survival of linear DNA, whereas complementation with full‑length or a catalytically inactive TelN restores viability, consistent with TelN playing a non‑enzymatic capping function.
In vitro, TelN binds hairpin substrates with moderate affinity and protects them from the nuclease activity of the Mre11/Rad50 complex. The authors propose that TelN originated as an early, sequence‑specific barrier against MR‑mediated DNA end processing, establishing fundamental principles of telomere protection that persist from bacteria to eukaryotes.
Major comments:
The manuscript convincingly shows that TelN can functionally block the Mre11‑Rad50 (MR) nuclease on a hair‑pin DNA end in a sequence specific manner (suggesting a physical interaction), but it doesn't directly demonstrate this. A simple pull-down or equilibrium binding method would useful in proving a physical interaction.
The MR complex requires ATP hydrolysis for resection of DNA ends. It would be a nice addition to the manuscript if the effect of TelN of Rad50 ATPase activity was tested.
The bar plot on Fig 3B indicates that the experiments are performed in triplicate. The statistical significance of the differences between conditions should be determined. The same general comment could be made regarding the quantification of the polyacrylamide gels - how reproducible are these values?
Minor comments:
A better explanation of how the gels were quantified should be provided. Were the products included in the analysis, or was it just the decrease in the substrate band that was measured?
I felt like the Results jump rather abruptly from B. subtilis chromosome assays to E. coli plasmid experiments. Maybe the addition of a few linking sentences would improve this transition.
A comment on the stoichiometry of TelN and genome ends during phage replication would be useful.
General assessment:
Strengths: A nice combination of genetics and biochemistry convincingly demonstrates that TelN protects linear chromosomes/replicons from MR-dependent degradation independent of its cleavage-ligase activity. It does this by binding to the hairpin DNA ends in a sequence specific fashion and the species specificity suggests a direct physical interaction, which likely inhibits the nuclease activity of the MR complex
Limitations: The lack of characterization of the putative physical interaction between TelN and the MR complex is considered a weakness.
Advance: The manuscript fills in a mechanistic gap between protelomerase‑mediated telomere formation and maintenance by demonstrating a protective/capping role. This is the first quantitative analysis of DNA-end protection from MR nuclease activity by TelN.
Audience: Readers interested in bacterial chromosome biology, DNA repair, the parallels to eukaryotic shelterin will be interesting to the broader telomere and genome‑stability communities.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The paper is well-presented and well-written throughout. The paper shows convincingly that TelN protects hairpin DNA ends from the activity of SbcCD, presumably providing a protection mechanism for N15 phage DNA in vivo. Furthermore, this protection activity is shown not to require the catalytic (resolvase) activity of TelN, nor its poorly characterised C-terminal domain. The paper also suggests that this inhibition acts both at the level of competition for the DNA hairpin end and at the level of a direct protein:protein interaction between TelN and MR. An (acknowledged) weakness is that there is no real insight into the protein:protein interaction suggested by the experiments shown in Figure 5. Ideally, the protein:protein interaction interface would be identified and mutations in this interface would be shown to reduce hairpin protection.
Specific comments/questions
(1) What pathway (in vivo) leads to inactivation of linear hairpin DNA - one suspects that cleavage by SbcCD at the hairpins is probably not the full story. Presumably SbcCD cleavage facilitates further processing by other long range resection systems such as RecBCD, Exo1, RecQ/J etc. Would it be appropriate to view the hairpin as an adaption to protect against these nucleases, which then must be complemented with a mechanism to suppress SbcCD?
(2) Section starting "Direct inhibition of MR by TelN in vitro". What is the word direct supposed to convey here? To me it suggests that the inhibition is via direct interaction of TelN with MR (rather than, for example, a result of competition for the hairpin DNA end) which is not shown here. Suggest either defining or removing the word direct. This point gains more importance considering that differentiating between inhibition mechanisms becomes a focus of later parts of the paper.
(3) Figure 2B - Why no control lane without MR? - this is a basic control to show that he degradation we are seeing in the absence of TelN is MR-dependent. Formally, as shown, the degradation could be caused by the ATP stock.
(4) Why not use B. subtilis SbcCD for the species specificity experiment? Also, is it not surprising that TelN yielded zero protection against MRX given that the DNA sequence specificity experiments above suggest competition for DNA substrate is part of the inhibition mechanism?
(5) If the authors felt it appropriate, I thought there was scope for further discussion/introductory material. There are strong parallels here with mechanisms used by phage to protect themselves from the activities of RecBCD, which include both proteins that protect DNA ends like T4 gene 2, we well as proteins that bind directly to RecBCD to inactivate it like lambda Gam. As such, the work here will appeal as much to those interested in bacterial defence systems / phage:host interactions as it does to those interested in telomere biology. Especially significant is the inhibition of DNA end processing factors by lambda Gam since this protein is reported to interact with both RecBCD and SbcCD (PMID: 2531105).
(6) Just a gripe really: it seems to be 'de rigeur' at the moment to re-name bacterial proteins for their human orthologues, presumably to elevate the perceived importance of the work(?), but it is not a practice I think is terribly helpful as it causes issues when searching literature. Minimally it would be great if the authors could ensure they add SbcCD as a keyword for search purposes.
Referee cross-commenting
I have nothing to add. The reviewers comments are all broadly positive and and consistent.
This is an excellent paper unveiling a phage encoded "counter-defence" mechanism designed to protect phage DNA from degradation. It will be of special interest to those studying telomere biology of phage:host interactions.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This study addresses how the bacterial telomere protein TelN protect telomere ends against the action of the Mre11-Rad50 nuclease (MR). This protection is essential for the stability of hairpin-ended linear plasmid and chromosomes in bacteria but had not been explored before. The authors demonstrates that TelN is necessary and sufficient to block MR-dependent DNA cleavage when bound to its specific telomere sequence. By combining elegant genetics and biochemical approaches, it convincingly shows that TelN-dependent inhibition likely involves a specific interaction between TelN and the MR complex. The manuscript is well written, easy to read and focused on the relevant information. The claims and the conclusions are supported by the data. There is no over-interpretation.
Comments:
Referee cross-commenting
Perfect for me. It seems that there is a consensus.
This pioneering study provides a very strong basis for a new understanding of telomeres in bacteria and offers fascinating evolutionary perspectives when compared to similar mechanisms active at telomeres in eukaryotic cells.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
We are very grateful for the positive feedback from all three reviewers. Below, we address each point in detail and outline proposed experiments and revision plans, with changes indicated by an underscore.
__Reviewer #1 (Evidence, reproducibility and clarity (Required)):
In this paper "Magnesium depletion unleashes two unusual modes of colistin resistance with different fitness costs," the authors examine how Pseudomonas aeruginosa evolves resistance to colistin, a last-resort antibiotic for multidrug-resistant Gram-negative infections. Although colistin resistance is a major clinical challenge, its underlying mechanisms, particularly under nutrient-limited conditions typical of infections, are not fully understood. The study shows that under low magnesium (Mg²_⁺_) conditions-mimicking infection or biofilm stress-P. aeruginosa can develop colistin resistance via two distinct genetic pathways, each with unique fitness costs. The first involves mutations in genes such as htrB2 and lpxO2, granting strong resistance but compromising the outer membrane and increasing susceptibility to other antibiotics. The second involves regulatory mutations (e.g., in the oprH/phoP/phoQ promoter) that confer resistance with minimal membrane defects and generally lower fitness costs. These resistance strategies lead to different trade-offs: membrane-compromising mutations reduce bacterial fitness without colistin, while regulatory mutations typically avoid these penalties, with context-dependent effects. The study underscores clinical relevance, noting that in infections-such as in cystic fibrosis-other microbes like Candida albicans may deplete magnesium, indirectly promoting resistance evolution. Overall, this work offers important insights into antibiotic resistance in nutrient-stressed, polymicrobial environments, highlighting how magnesium availability shapes resistance evolution and fitness costs. The findings suggest new avenues for therapeutic intervention and call for a reevaluation of antibiotic strategies in nutrient-competitive infection settings.
Work is timely and important. Colistin resistance represents an urgent threat as colistin is a last-resort antibiotic used against multidrug-resistant Gram-negative pathogens. Insights into mechanisms evolving under nutrient limitation are highly relevant given the prevalence of such environmental conditions during infection and microbial biofilm growth. The study reveals two previously uncharacterized pathways to colistin resistance in P. aeruginosa triggered by magnesium (Mg²_⁺_) depletion, each with distinct genetic signatures and trade-offs. This finding directly impacts the understanding of polymicrobial infection dynamics, especially where magnesium sequestration by fungi/ or other microbes may occur. The identification of fitness costs and pleiotropic effects associated with specific resistance mutations provides crucial guidance for clinicians considering antibiotic stewardship and combination therapy strategies.
__
We thank the reviewer for their summary of our study and its potential impact.
__Drawbacks
• Experimental scope: While the study is comprehensive for P. aeruginosa, the broader applicability to other Gram-negative pathogens is not directly tested.__
In our revision, we now explicitly point out that the magnesium limitation we have observed broadly applies to Gram-negative bacteria, as we demonstrated in our previous PLOS Biology paper. Therefore, we expect the same themes (and even genes, which are broadly conserved) to apply to Gram-negative bacteria in general. However, a full-fledged experimental study of other Gram-negative pathogens is outside the scope of our current study, which required a 90-day experimental evolution.
__Strengths
• Experimental evolution: This work uses laboratory evolution under controlled Mg²_⁺_-limited conditions to simulate selection pressures relevant to infection microenvironments. • Genetics: Systematic identification and functional validation of key mutations-particularly in htrB2, lpxO2, and the oprH/phoP/phoQ promoter-give mechanistic depth to the findings. • Two distinct resistance modes: Evidence for (i) one pathway leading to colistin resistance via htrB2 mutations, resulting in high resistance but significant membrane integrity loss and increased susceptibility to other antibiotics. (ii) a second pathway providing resistance without compromising membrane integrity, highlighting evolutionary flexibility and ecological implications. • Fitness assessments: measurement of the costs associated with each resistance strategy, both in terms of membrane integrity and susceptibility to other agents. • Relevance: Connection to natural scenarios, such as magnesium sequestration by fungi (e.g. Candida albicans) in polymicrobial environments, underscores the ecological and clinical significance. • This manuscript is well written with clearly logical hypothesis testing__
We thank the reviewer for their appraisal, especially for recognizing the rigor and broader biological implications of our study.
__Drawbacks
• Experimental scope: While the study is comprehensive for P. aeruginosa, the broader applicability to other Gram-negative pathogens is not directly tested.__
We agree with the reviewer's point about broader applicability in other Gram-negative bacteria, as many of the lipid A biosynthesis genes are conserved among diverse bacterial lineages. We will include this point in our revised Discussion to suggest relevance to other Gram-negative bacteria:
"We previously showed that magnesium sequestration by fungi applies not only to P. aeruginosa but to other Gram-negative bacteria as well (ref). Our current study lays a foundation for developing evolution-guided strategies to combat multidrug-resistant P. aeruginosa and other Gram-negative bacteria that can also acquire colistin resistance. Since many other antibiotic mechanisms are similarly dependent on metal ions (refs), our work suggests that nutritional competition for metal ions may alter initial antibiotic resistance in Gram-negative bacteria and potentiate new evolutionary pathways of antibiotic resistance."
__ Mechanistic depth: Some inferred mechanisms (e.g., the precise molecular impact of late-occurring adaptive mutations) merit deeper biochemical analysis.__ We will emphasize in our Revision that the MS data of endpoint clones and triple mutants reveal that their lipid A structures are identical. This suggests that the role of other late-occurring mutations in enhancing resistance is likely through lipid A-independent pathways.
__ Results Lines 414- 423: While correlation is most what makes sense for some drugs, causality is implied (membrane defects increase susceptibility), but could be strengthened by directly measuring antibiotic uptake (e.g., fluorescence) or membrane permeability for these 3 antibiotics.__ We thank the reviewer for highlighting the issue of causality. For the three antibiotics tested, the most direct way to measure their effect is by measuring their impact on bacterial growth directly, which is what we have done. Our membrane permeability assay using NpN uptake operates under the same conditions suggested by the reviewer and directly measures molecular uptake. Moreover, only fluorescently labeled vancomycin is commercially available among the three antibiotics tested. Since it binds to the cell wall, its utility to measure membrane defects is more limited than the NpN assay we have already used. However, in response to this comment, we will make clear in our revision that we infer that increased susceptibility to other antibiotics is due to their increased membrane permeability.
__ o Effect is mild and mostly not significant. It is also not clear whether authors only tested a handful of mutants shown in Fig. 7B-D or whether other clones were also tested. The sample of endpoints (P2, P5, P8) covers well-characterized lineages, but additional evolved clones or a broader panel could boost generality about other antibiotics. The authors note "significantly lower MICs" statistical treatment is implied; explicit statistical values and replicate numbers should be given in the text or figures.__
We slightly disagree with the reviewer that the results are not significant. Even two-to-three-fold differences in MICs translate to large differences in microbial competition. These three endpoint clones are representative of all eight evolved strains after 90-day evolution experiments. Moreover, we will emphasize in the Revision that we have tested all the mutations found in the endpoint clones; we know what these are from whole genome sequencing of multiple endpoint clones. In addition, we will explicitly state the p-value in the legend of Figure 7.
__ The structural or physiological nature of "mild" vs. "severe" membrane defects could be better defined/quantified.__ Although we agree with the reviewer's suggestion, the variability of the SEM assay makes the classification of membrane defects based on cell morphology hard to quantify. We therefore only use the SEM images as representative of the various defects observed. For a more quantitative assay of the membrane defects, we instead rely on the standard NpN uptake assay to quantify membrane permeability as a quantifiable readout for membrane defects.
__ Quantitative limits: Authors should add in the discussion that statistical robustness could be strengthened-for example, by including longer-term evolutionary predictions.__ We are not sure what the reviewer means and so cannot address this point completely. We ask the reviewer to rephrase this point, and we will address it to the best of our abilities.
__ in vivo relevance: While the ecological context is discussed, direct in vivo confirmation (e.g., in animal infection models) of the observed resistance trajectories would increase translational impact and relevance.__ We agree with the reviewer's point. However, it is not trivial to directly perform evolution experiments of microbes in animal models. There are only a handful of labs worldwide that have working CF-relevant animal models. However, the colistin resistance mutations we identified provide a tool to look deeper into how colistin-resistant P. aeruginosa can evolve in vivo.
__ Some sections are repetitive or overly detailed; condense where possible (especially on mutation lists and background for each claim).__
We will condense our manuscript as the reviewer suggested in our revision. Adding a graphical summary as suggested will also allow us to be more succinct in our description.
__Other comments
• Authors should provide clarification on how the Mg²_⁺_ concentrations used in vitro compare to those found in clinically relevant infection settings. This would be helpful to enhance significance.__
We thank the reviewer for raising this good point. Based on our previous work, we know the Mg2+ levels in our model (0.3-0.45mM) are within the physiological range of Mg2+ in infection settings (0.1-0.8mM). We will highlight this point in the introduction.
We will include the details of our statistical tests in each panel of figures both in the main text and the supplement.
We will name each of the particular mutations tested to be specific about the nature of all the evolved mutations in our figure legends.
__ The manuscript could benefit from a graphical summary illustrating the two distinct evolutionary pathways and their respective fitness landscapes.__ We thank the reviewer for this suggestion to enhance the clarity of our work. We will make a new graphical summary highlighting two different evolutionary pathways as a new figure.
__ A brief discussion of therapeutic implications-such as combining colistin with agents that target membrane integrity-would help bridge the gap from mechanism to clinical management.__ In our discussion, we have suggested that collateral sensitivity (line 446-453) and PhoPQ kinase inhibitors (line 512-515) could be exploited to combat colistin resistance. To make this point more clearly, we will slightly expand our Discussion to include the therapeutic implications of our study.
__ Additional discussion on whether the fitness costs are reversible or can be compensated by further adaptation would be valuable for long-term dynamics.__ We thank the reviewer for raising this interesting point. The evolution trajectory of P8 suggests that fitness costs can be compensated by later-occurring mutations during evolution. We will further discuss this point to highlight the importance of understanding the mutational dynamics of antibiotic resistance evolution.
__ It would be valuable for the authors to comment on, or further analyze, whether there is a direct association between specific fitness costs and sensitivity to other antibiotics. Such information could inform on evolutionary constraints and possible trade-offs relevant to clinical settings.__
We will include a supplemental figure showing the correlation between fitness costs and antibiotic susceptibility for P2, P5, and P8.
__ Main figures and support for claims
The main and supplementary figures comprehensively illustrate the evolutionary trajectories, genetic bases, and phenotypic outcomes associated with colistin resistance under magnesium depletion in P. aeruginosa. The figures effectively detail: • Genetic pathways involved including the experimental evolution design (colistin selection under Mg²_⁺_ depletion), whole-genome sequencing results, and timelines of observed mutations (e.g., in htrB2, lpxO2, oprH/phoP/phoQ promoter, PA4824). • Phenotypes and biochemical analyses such as lipid A structure (via mass spectrometry), minimum inhibitory concentration (MIC) assays, and epistasis analyses between mutations are depicted. • Fitness trade-offs are demonstrated using bacterial survival, membrane integrity (e.g., scanning electron microscopy images), membrane permeability assays (NPN uptake), and competitive fitness assays. • Mechanistic claims about the necessity of early mutations, the requirement of the PhoPQ pathway at different evolutionary stages, and the fitness cost imposed by certain resistance mutations. To further enhance the rigor and clarity of the manuscript, the authors should implement the following improvements: • Labelling consistency: In some instances, figure legends could provide more granular detail about specific mutations (e.g., positions of amino acid changes). • Graphical summary: A schematic summary figure that visually integrates the three main evolutionary resistance trajectories, the mutational order, corresponding lipid A changes, and fitness costs, would enhance readability. • Replicates: Plots should more thoroughly indicate the number of replicates and show individual data points (not just means {plus minus} SD), add number of replicates in each experiment. • Supplementary: figures referenced in the text (e.g., lipid A structures or mutation reversion outcomes) should be made more prominent or better cross-referenced from the main results section. Authors should highlight when supplementary data provide critical functional confirmation (e.g., confirming mutation function or fitness reversal).__
We thank the reviewer for their appreciation of our work and constructive feedback.
__Statistics
The authors have appropriately incorporated statistical analyses throughout the figures. To enhance the robustness and credibility of their findings, authors should also cross-check • Tests in legends: Every figure and supplementary figure should clearly state the type of statistical test used, how many biological replicates, and any corrections for multiple comparisons.__
As mentioned above, we will provide more details about the statistical tests of each panel.
__ Effect sizes: Where appropriate, reporting effect sizes-rather than just p values-would contextualize the biological impact.__ We agree with the reviewer; we will mention the magnitude of MIC changes in the corresponding figure legends.
__ Raw data accessibility: For full transparency, consider sharing underlying raw data and analysis scripts.
__ We will provide the raw data of each panel.
__Overall, the main and supplementary figures effectively illustrate and substantiate the key claims-particularly the alternative molecular pathways, phenotypic trade-offs, and the role of environmental magnesium in mediating colistin resistance. Statistical analysis is generally robust and appropriately presented throughout, though improvements could include more explicit reporting, additional controls, and accessible raw data. The visual and quantitative data in the figures provide support for the authors' conclusions about the evolution of antibiotic resistance under nutrient limitation in microbial environments. Understanding these alternative pathways is important for designing better treatment strategies and for predicting how resistance might evolve under varying clinical and environmental conditions.
__
We thank the reviewer for their positive assessment.
__ Reviewer #1 (Significance (Required)):
Overall, this work offers important insights into antibiotic resistance in nutrient-stressed, polymicrobial environments, highlighting how magnesium availability shapes resistance evolution and fitness costs. The findings suggest new avenues for therapeutic intervention and call for a reevaluation of antibiotic strategies in nutrient-competitive infection settings.__
We sincerely thank the reviewer for constructive and thoughtful feedback and the acknowledgement of our figure presentation and experimental design. We feel very encouraged by the reviewer's perspective that our study provides unique insights into resistance evolution in polymicrobial environments and may inform therapeutic strategies.
__My expertise: Gut microbiome, gut microbiota resilience, ecology, and evolution in microbial communities, antimicrobial resistance, high-throughput drug-bacteria interactions
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
Summary: The paper by Hsieh and colleagues unravels the molecular basis of colistin resistance in Pseudomonas aeruginosa under low magnesium (Mg2+) conditions. Colistin is a last resort antibiotic that compromises bacterial cell wall integrity. Bacteria can respond (phenotypically and genotypically) to colistin by modifying membrane-anchored lipopolysaccharides. Mg2+ depletion can trigger similar responses. In their study, Hsieh et al. find that Mg2+ depletion (induced by a co-infecting fungal pathogen, Candida albicans) leads to evolutionary trajectories and resistance mechanisms that differ from those observed under Mg-rich conditions. The authors conducted a series of detailed genetic, chemical and fitness-based experiments to elucidate the molecular, physiological and evolutionary basis of these new resistance mechanisms.__
We thank the reviewer for their summary of our study.__
Major comments: __ 1. The authors reconstituted key mutations observed during experimental evolution in the ancestral background. Moreover, they took clones from the final stage of the evolution experiment and restored the ancestral state of the mutated genes. This dual approach is extremely strong and allows to decipher the causal effects of colistin resistance. I like to applaud the authors for this rigorous approach.
We thank the reviewer's appreciation about the rigor and comprehensive analyses of our study.
2. I understand that this work focusses on evolved mutants isolated from a previous experiment. The focus is on Mg2+ limitation. However, it would still have been nice to include a characterised colistin resistent strain featuring more standard resistance mechanisms. How different would such a strain be in the analyses shown in Fig. 3? Would morphological changes (Fig. 5A), fitness trade-offs (Fig. 6) and collateral sensitivity (Fig. 7) also occur in such a mutant. I do not regard it as imperative to include data from such a strain. But putting the new data into context (at least in the discussion) would clearly increase the overall impact of this work.
We thank the reviewer for raising this fascinating and vital point. We will address the point in our Revision using the monoculture (high Mg2+) evolved strains, which acquired many known mutations for colistin resistance, as our reference. We will provide a supplemental figure about the membrane permeability, fitness costs, and collateral sensitivity of monoculture evolved strains. We will also contrast their difference from co-culture evolved strains in the revised Discussion.__
We thank the reviewer for pointing out this important reference. We will include this reference and its findings in the Discussion.
__Minor comments:
We thank the reviewer for this suggestion. Figures 1A and 1B summarize the previous paper; all other panels are new data. We will make this clear in the revised text and figure legend.
5. MIC-data (e.g. Fig. 2) come in discrete categories (based on the underlying dilution series). This comes with some challenges for statistical analysis. First, linear models like ANOVAs are based on normally distributed residuals. This is violated with discrete data distributions. Second, there is often no within-treatment variation (e.g., Fig. 2B), which makes statistical analyses obsolete. These points need to be addressed. Moreover, how is it possible to have subtle variations in MIC (e.g., Fig. 2A, P2 endpoint clone) with classic dilution series (as indicated on the y-axis, 128, 256, 512)? Please explain.
We agree with the reviewer that statistical analysis of MIC data is not straightforward. ANOVAs are not well-suited for this type of discrete data, and the lack of variation within replicates reduces the power of non-parametric tests such as the Mann-Whitney U test. To improve the statistical reporting of MIC data, we will apply non-parametric tests and include effect size measurements, as recommended by Reviewer 1.
Moreover, the design of dilution series may underestimate the true nature of antibiotic susceptibility. To address these issues, we have also performed survival assays to assess colistin resistance in both the endpoint and reversion strains; we will also include statistics to assess the significance of their different survival frequencies.
We thank the reviewer for highlighting the point about subtle variations in a classical dilution series. Our endpoint strains grew robustly in media containing 192 μg/mL colistin-the highest concentration used in our evolution experiment. To more accurately determine and compare their maximum MICs, we expanded the colistin concentration range using finer fold increases (1.5×, 2×, 2.5×, 3×, 3.5×, and 4×) from 192 to 768 μg/mL. We will update these details in the Materials & Methods.
__ Lines 264-269. This analysis focusses on enzyme impairment. However, mutations could also change enzyme activity. Could any of these mutations have such an effect?__
The answer is "yes". As evolved strains with lpxA mutation still have lipid A, we suspect this mutation does not altogether abolish lipid A synthesis. However, this mutation could affect the amount of lipid A or change enzyme specificity. These are interesting ideas for further investigation, but they fall beyond the scope of our current study. We will, however, include the requested detail in the discussion.
__ Figure 5A. Some arrows seem to be out of place and point at void spaces. Please check.__
We thank the reviewer for pointing out this error, which we will correct.
8. The use of polymyxin B is not well justified (Fig. 5 and Fig. S13). Did the authors aim to test whether there is cross-resistance to other antimicrobial peptides?
We will more clearly justify our choice of using polymyxin B for directly assaying binding of polymyxin antibiotics to bacterial cells using fluorescence-labeled polymyxins, since no such reagents exist for colistin and since previous studies (including ours) have shown similarity of susceptibility to colistin and polymyxin B:
"Although P2 and P5 endpoint clones have more permeable membranes, they exhibited greater resistance to polymyxin antibiotics, including colistin (polymyxin E) (Fig. 5D), and polymyxin B (Fig. S13A) than WT cells. To investigate how membrane-compromised cells gain increased resistance to antibiotics that target the outer membrane, we used dansyl-labeled polymyxin B [51] to quantify the binding of polymyxins to P. aeruginosa; dansyl-labeled polymyxin fluoresces upon binding the hydrophobic portion of bacterial membranes. We used polymyxin B binding as a surrogate for how bacterial cells bind to all polymyxin antibiotics, including colistin."
__ Line 564. Please indicate the dilution factor used.__
Thank you for pointing out this inadvertent omission. We will update our Materials & Methods accordingly, as in response to the Reviewer 2's comment 5.
__Reviewer #2 (Significance (Required)):
This is a very strong and well designed study. It provides novel and relevant insights into the resistance mechanisms against an important last resort antibiotic.__
We sincerely thank the reviewer for their thoughtful summary and generous evaluation of our work.
__Reviewer #3 (Evidence, reproducibility and clarity (Required)):
This manuscript reports on biologically interesting and clinically-relevant findings, that upon passaging in the presence of spent media from C. albicans, P. aeruginosa develops resistance to colistin through lipid A modifications. The authors thoroughly characterize novel lipid A structures seen in their resistant mutants, and test a variety of genetically constructed mutants to determine the contributions of specific mutant alleles to resistance.__
We thank the reviewer for the appreciation of our experimental design and comprehensive genetic and biochemical analyses of our evolved strains.
However, additional experiments are needed to demonstrate the specific role and necessity of the lipid modifications for colistin resistance.
We are also grateful for the reviewer's feedback and constructive criticisms to improve the clarity and impact of our manuscript. We have listed detailed responses to the reviewer below.
Additionally, as Reviewer 2 pointed out, both mutation reconstruction and reversion experiments are required for understanding the roles of each mutation and interactions among different mutations in contributing to resistance. Combining all the results of htrB2 and lpxO2 mutations in these two orthogonal genetic experiments, it is the synergistic interactions among these mutations that lead to enhanced resistance after evolution. This explains why we saw genetic background effects of htrB2 mutation (P2 vs P5) and why each single mutation is required for resistance but doesn't contribute to resistance significantly by itself.
- In P8, the effect of a single lpxA mutation is not tested. Further, the resistance of a P-oprH + lpxA mutant is the same as that of just the P-oprH mutant, indicating that the lpxA mutation likely does not directly alter colistin resistance. It is possible that mutations in lpxA were selected to compensate for fitness defects resulting from the other mutations, or for adaptation to some other component of the media conditions.
This is an excellent suggestion. We will assess the MIC and fitness of reconstructed strains with the lpxA mutation to update the role of this mutation.
- While reversion of the htrB2 and lpxO2 mutations do lead to ~3-4x reduced resistance in P5 indicating some contribution of these mutations, it is specific to this population, and thus not clear whether it is due to the specific lipid A modifications (some of which are seen in the other populations too). A specific combination of lipid A modifications may confer colistin resistance, but this needs to be demonstrated by generating just those clean deletion mutants and showing an effect on resistance.
In response to this comment and comment 1, we will make lpxO2 deletions in WT, the triple mutant and the endpoint clone of P5 to test colistin resistance. However, our results of reverting single htrB2 or lpxO2 mutation to WT are robust and use two independent assays, including the standard MIC test and colistin survival assay. So, we are confident that each mutation is necessary for enhancing colistin resistance.
__ Overall, given the high levels of colistin resistance still exhibited by single mutant revertants (Fig. S3) and the absence of double or triple revertants, it is hard to come to any conclusions regarding causality. This is especially the case for P8 but also true of P2 and P5. What are the other mutations in these populations, and what role do they play in colistin resistance?__
We respectfully disagree with the reviewer on this point. One point that we have made and will re-emphasize in our Revision is that we have assayed all the mutations in these populations; this is one of the advantages of our experimental evolution and genome sequencing strategy. All the mutations that could play a role in colistin resistance have therefore been tested. Furthermore, due to genetic epistasis of mutations in different evolutionary lineages, we do not necessarily expect that a single revertant would altogether abolish colistin resistance, as has been demonstrated in several previous studies. As Reviewer 2 pointed out, combining mutation reconstruction and reversion is the best way to establish causality, and we have done so. Therefore, it is not correct to say that we cannot come to 'any conclusions regarding causality'.
__ Figure 4 is titled "The PhoPQ pathway synergizes with early-arising mutations to confer colistin resistance.", but instead what this figure shows is that the mutation upstream of oprH increases PhoP activity. I'm not sure what the synergy here is. The same is true for the section starting on line 276. Further, the first sentence of that section states "We next investigated why the mutations conferring robust colistin resistance in low Mg2+ conditions are not observed in Mg2+ replete conditions.". However, there are no experiments there testing whether the mutations conferred resistance in Mg2+ conditions, instead the authors just test whether the mutations they are studying increase PhoP activity, and require PhoPQ to confer resistance.__
We thank the reviewer for raising this point. We apologize for the unclear writing. We will use this opportunity to improve the clarity of this section by rewriting it to focus on two points: 1. Evolved resistance is PhoPQ-dependent, instead of PmrAB-dependent. 2. Two lineages evolved enhanced resistance by boosting PhoPQ activity in both high and low Mg2+ conditions. We will also remove the statement highlighted by the reviewer from this section that obfuscates the motivation of this section. We feel this approach will more clearly show how lipid A-related mutations contribute to resistance in low Mg2+.
__ The authors claim that the identified mutations did not appear in the high magnesium conditions because they had a fitness cost under those conditions, but figure 6A shows that the evolved strains have fitness costs in low magnesium conditions as well. Further, the authors suggest that because the studied mutations act via increased PhoPQ activity, they do not lead to resistance under high magnesium conditions (lines 376-379). However, the increased PhoPQ activity is mediated by the P-oprH mutation in the isolates which likely increases PhoPQ activity even in high magnesium conditions. Overall, it is not clear why the mutations in the low magnesium condition were not selected for under high magnesium conditions.__
The reviewer is correct about the fitness cost in high Mg2+ and low Mg2+ conditions. These fitness experiments were carried out in the absence of colistin, which explains the finding that there are fitness defects in both conditions. As is well known, evolution for antibiotic resistance will ultimately select for resistant mutants, despite their fitness costs. In contrast, colistin MIC of these endpoint strains in high Mg2+ conditions was still much lower than the colistin concentration we applied during evolution (Fig. S15), indicating it is much less likely for these mutations to be selected for in high Mg2+. We will clarify this point in our revised Results and Discussion.
We agree with the reviewer about the P-oprH mutations (PhoPQ expression) and will note that, unlike the other mutations, it is not clear why these emerge only in the low Mg2+ condition.
__ The authors used C. albicans spent BHI media as their low magnesium condition, but this condition has a lot of other C. albicans metabolites that may be affecting the results. It is possible that what the authors are observing is not related to magnesium at all, and the authors should test the phenotypes in normal BHI medium depleted for magnesium or some defined medium where magnesium levels can be controlled.__
We thank the reviewer for mentioning this important point. In our prior PLOS Biology paper (https://doi.org/10.1371/journal.pbio.3002694.g005), we demonstrated that supplementing Mg2+ in evolved co-culture populations reduces colistin resistance, suggesting this evolved resistance is Mg2+ dependent. We also know that the MIC of our endpoint strains in C. albicans-spent BHI with supplemented Mg2+ (MIC of all three endpoint clones is less than 48 mg/mL colistin) is much lower than in C. albicans-spent BHI. We will mention this detail in the paper and include the data in our revision if the reviewer and editor require it.
Other comments: - The authors use MIC assays as well as % survival to measure resistance against colistin, and sometimes use both in the same figure (e.g. Figure 2). This makes direct comparisons difficult. It would be better to consistently use one assay, preferably the MIC, at least in all the main figures. If the survival data needs to be included, it could go in the supplementary figures.
We thank the reviewer for this suggestion. We will move the MIC data of mutation-reversion strains to the main Fig. 2D-F.
- While the mutations seen in the low and high magnesium conditions were shown in the previous manuscript, given the extensive dissection here, it would be useful for readers if the authors gave some details about the serial passaging and evolution experiment, identification of mutations, and some mention of what mutations were seen in high Mg populations.
We will add these details in the introduction.
- Given that oprH is present in an operon, it would be more accurate to call that mutation as being in the promoter of the oprH-phoP-phoQ operon rather than it being an oprH mutation (at least in the text, e.g. lines 127-129).
We agree. We will change this as the reviewer requested.
- Unlike what is stated on lines 287-290, deletion of oprH in P2 leads to a greater than 2x reduction in colistin MIC, suggesting that OprH is playing a role (albeit a smaller role than phoP) - Line 50 has a typo, remove "160". - Line 122: Specify which Pa and Ca strain backgrounds were used. - Line 132: Were representative isolates derived from terminal passages? This should be defined.
We will change these points according to the reviewer's suggestions; we thank them for these suggestions.
- Line 215-219: It is interesting that Pa WT grown in spent medium additionally results in lipid A that is hexa-acylated. Is this sufficient to alter colistin resistance on its own?
We find that WT PAO1 in low Mg2+ conditions has PagP-mediated acylation, which can slightly increase colistin resistance, but not to the extent of resistance as our evolved strains.
- It would be useful to see a PCA plot for the samples shown in figures S6 and S7.
We will include such a plot in Figures S6 and S7
- Fig. S11: What are the colistin MICs of pmrA and phoP deletions in the WT background?
MIC of pmrA and phoP deletions in WT is 1.5ug/mL. We will include these data in the Revision.
- Instead of qualitative data, can the authors quantify cell length and perhaps some measure of cell shape (instead of just showing images in Fig. 5A and S12).
We thank the reviewer for raising this point. A similar comment was raised by Reviewer 1. As it's challenging to quantify membrane changes from the morphological data obtained through SEM (a point which we will now clarify in our Revision), we used a quantifiable NpN uptake assay to quantify membrane defects of our evolved strains.
- What is the WT MIC in high magnesium conditions? Please show that in Fig. S15.
We will include this detail in Fig. S15
- I am not an expert in lipid modifications and structures, but in figure S5, P2 and P4 show high peaks with lower m/z that seem specific to low magnesium conditions, but they are not labeled or discussed. What are these peaks?
We thank the reviewer for bringing up this concern. The unlabeled lipids in these spectra are cardiolipin, not lipid A. These peaks are present in all the samples, and the reason they appear larger in the P1 and P4 low magnesium conditions is that both spectra are scaled to the relative intensity of one another. It is important to note that MALDI-TOF MS is not a quantitative technique, and the relative intensity of the peak heights between two samples should not be used to compare the amounts of lipids in one sample versus another. Therefore, we cannot say that these lipids are present in greater quantities in low magnesium conditions versus high magnesium conditions.
- Lines 357-358 state that "mutant cells minimally bind polymyxin B (Fig. S13B)", but the figure shows increased binding compared to the WT. The legend of the figure also says something similar. Are the phoP pmrA mutants expected to bind more polymyxin B because they can't modify lipid A?
We thank the reviewer for pointing out this substantial error. We will change 'minimally bind' to 'demonstrate increased binding'.
- Given the fitness defects in just regular medium, is the data shown in Figure 7 specific collateral sensitivity to the antibiotics tested? Are there other conditions where P2 and P5 do not show increased sensitivity?
These are all the antibiotics we have tested. It is conceivable that P2 and P5 might not show increased sensitivity to other antibiotics that use the same mode of action as colistin or polymyxin B.
__Reviewer #3 (Significance (Required)):
This study aims to dissect novel mechanisms of colistin resistance in P. aeruginosa that arise upon passaging in C. albicans spent media. While the authors identify novel lipid A modifications associated with the evolved strains, the significance of the modifications for resistance, and the mechanisms for why these evolutionary trajectories were not selected for in high magnesium are not clear from the data presented.__
We thank the reviewer for recognizing the integrity of our work and for the constructive feedback on improving the clarity of our writing. We understand that some concerns may stem from a lack of clarity in our original submission, but that additional genetic experiments are necessary. We have already identified all mutations that arose independently across different lineages and characterized their contributions to resistance, which we believe supports a robust inference of causality. To strengthen our conclusions, we will incorporate additional experiments, including htrB2 deletion, lpxO2 deletion, and lpxA mutation, to better dissect the roles of these genes and mutations in colistin resistance. We hope this revision plan will ameliorate the reviewer's concerns.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript reports on biologically interesting and clinically-relevant findings, that upon passaging in the presence of spent media from C. albicans, P. aeruginosa develops resistance to colistin through lipid A modifications. The authors thoroughly characterize novel lipid A structures seen in their resistant mutants, and test a variety of genetically constructed mutants to determine the contributions of specific mutant alleles to resistance. However, additional experiments are needed to demonstrate the specific role and necessity of the lipid modifications for colistin resistance.
Other comments:
This study aims to dissect novel mechanisms of colistin resistance in P. aeruginosa that arise upon passaging in C. albicans spent media. While the authors identify novel lipid A modifications associated with the evolved strains, the significance of the modifications for resistance, and the mechanisms for why these evolutionary trajectories were not selected for in high magnesium are not clear from the data presented.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary: The paper by Hsieh and colleagues unravels the molecular basis of colistin resistance in Pseudomonas aeruginosa under low magnesium (Mg2+) conditions. Colistin is a last resort antibiotic that compromises bacterial cell wall integrity. Bacteria can respond (phenotypically and genotypically) to colistin by modifying membrane-anchored lipopolysaccharides. Mg2+ depletion can trigger similar responses. In their study, Hsieh et al. find that Mg2+ depletion (induced by a co-infecting fungal pathogen, Candida albicans) leads to evolutionary trajectories and resistance mechanisms that differ from those observed under Mg-rich conditions. The authors conducted a series of detailed genetic, chemical and fitness-based experiments to elucidate the molecular, physiological and evolutionary basis of these new resistance mechanisms.
Major comments:
Minor comments:
This is a very strong and well designed study. It provides novel and relevant insights into the resistance mechanisms against an important last resort antibiotic.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this paper "Magnesium depletion unleashes two unusual modes of colistin resistance with different fitness costs," the authors examine how Pseudomonas aeruginosa evolves resistance to colistin, a last-resort antibiotic for multidrug-resistant Gram-negative infections. Although colistin resistance is a major clinical challenge, its underlying mechanisms, particularly under nutrient-limited conditions typical of infections, are not fully understood.
The study shows that under low magnesium (Mg²⁺) conditions-mimicking infection or biofilm stress-P. aeruginosa can develop colistin resistance via two distinct genetic pathways, each with unique fitness costs. The first involves mutations in genes such as htrB2 and lpxO2, granting strong resistance but compromising the outer membrane and increasing susceptibility to other antibiotics. The second involves regulatory mutations (e.g., in the oprH/phoP/phoQ promoter) that confer resistance with minimal membrane defects and generally lower fitness costs. These resistance strategies lead to different trade-offs: membrane-compromising mutations reduce bacterial fitness without colistin, while regulatory mutations typically avoid these penalties, with context-dependent effects. The study underscores clinical relevance, noting that in infections-such as in cystic fibrosis-other microbes like Candida albicans may deplete magnesium, indirectly promoting resistance evolution. Overall, this work offers important insights into antibiotic resistance in nutrient-stressed, polymicrobial environments, highlighting how magnesium availability shapes resistance evolution and fitness costs. The findings suggest new avenues for therapeutic intervention and call for a reevaluation of antibiotic strategies in nutrient-competitive infection settings.
Work is timely and important. Colistin resistance represents an urgent threat as colistin is a last-resort antibiotic used against multidrug-resistant Gram-negative pathogens. Insights into mechanisms evolving under nutrient limitation are highly relevant given the prevalence of such environmental conditions during infection and microbial biofilm growth. The study reveals two previously uncharacterized pathways to colistin resistance in P. aeruginosa triggered by magnesium (Mg²⁺) depletion, each with distinct genetic signatures and trade-offs. This finding directly impacts the understanding of polymicrobial infection dynamics, especially where magnesium sequestration by fungi/ or other microbes may occur. The identification of fitness costs and pleiotropic effects associated with specific resistance mutations provides crucial guidance for clinicians considering antibiotic stewardship and combination therapy strategies.
Strengths
Drawbacks
Other comments
Main figures and support for claims
The main and supplementary figures comprehensively illustrate the evolutionary trajectories, genetic bases, and phenotypic outcomes associated with colistin resistance under magnesium depletion in P. aeruginosa. The figures effectively detail: - Genetic pathways involved including the experimental evolution design (colistin selection under Mg²⁺ depletion), whole-genome sequencing results, and timelines of observed mutations (e.g., in htrB2, lpxO2, oprH/phoP/phoQ promoter, PA4824). - Phenotypes and biochemical analyses such as lipid A structure (via mass spectrometry), minimum inhibitory concentration (MIC) assays, and epistasis analyses between mutations are depicted. - Fitness trade-offs are demonstrated using bacterial survival, membrane integrity (e.g., scanning electron microscopy images), membrane permeability assays (NPN uptake), and competitive fitness assays. - Mechanistic claims about the necessity of early mutations, the requirement of the PhoPQ pathway at different evolutionary stages, and the fitness cost imposed by certain resistance mutations. To further enhance the rigor and clarity of the manuscript, the authors should implement the following improvements: - Labelling consistency: In some instances, figure legends could provide more granular detail about specific mutations (e.g., positions of amino acid changes). - Graphical summary: A schematic summary figure that visually integrates the three main evolutionary resistance trajectories, the mutational order, corresponding lipid A changes, and fitness costs, would enhance readability. - Replicates: Plots should more thoroughly indicate the number of replicates and show individual data points (not just means {plus minus} SD), add number of replicates in each experiment. - Supplementary: figures referenced in the text (e.g., lipid A structures or mutation reversion outcomes) should be made more prominent or better cross-referenced from the main results section. Authors should highlight when supplementary data provide critical functional confirmation (e.g., confirming mutation function or fitness reversal).
Statistics
The authors have appropriately incorporated statistical analyses throughout the figures. To enhance the robustness and credibility of their findings, authors should also cross-check - Tests in legends: Every figure and supplementary figure should clearly state the type of statistical test used, how many biological replicates, and any corrections for multiple comparisons. - Effect sizes: Where appropriate, reporting effect sizes-rather than just p values-would contextualize the biological impact. - Raw data accessibility: For full transparency, consider sharing underlying raw data and analysis scripts.
Overall, the main and supplementary figures effectively illustrate and substantiate the key claims-particularly the alternative molecular pathways, phenotypic trade-offs, and the role of environmental magnesium in mediating colistin resistance. Statistical analysis is generally robust and appropriately presented throughout, though improvements could include more explicit reporting, additional controls, and accessible raw data. The visual and quantitative data in the figures provide support for the authors' conclusions about the evolution of antibiotic resistance under nutrient limitation in microbial environments. Understanding these alternative pathways is important for designing better treatment strategies and for predicting how resistance might evolve under varying clinical and environmental conditions.
Overall, this work offers important insights into antibiotic resistance in nutrient-stressed, polymicrobial environments, highlighting how magnesium availability shapes resistance evolution and fitness costs. The findings suggest new avenues for therapeutic intervention and call for a reevaluation of antibiotic strategies in nutrient-competitive infection settings.
My expertise:
Gut microbiome, gut microbiota resilience, ecology, and evolution in microbial communities, antimicrobial resistance, high-throughput drug-bacteria interactions
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
*The authors have a longstanding focus and reputation on single cell sequencing technology development and application. In this current study, the authors developed a novel single-cell multi-omic assay termed "T-ChIC" so that to jointly profile the histone modifications along with the full-length transcriptome from the same single cells, analyzed the dynamic relationship between chromatin state and gene expression during zebrafish development and cell fate determination. In general, the assay works well, the data look convincing and conclusions are beneficial to the community. *
Thank you for your positive feedback.
*There are several single-cell methodologies all claim to co-profile chromatin modifications and gene expression from the same individual cell, such as CoTECH, Paired-tag and others. Although T-ChIC employs pA-Mnase and IVT to obtain these modalities from single cells which are different, could the author provide some direct comparisons among all these technologies to see whether T-ChIC outperforms? *
In a separate technical manuscript describing the application of T-ChIC in mouse cells (Zeller, Blotenburg et al 2024, bioRxiv, 2024.05. 09.593364), we have provided a direct comparison of data quality between T-ChIC and other single-cell methods for chromatin-RNA co-profiling (Please refer to Fig. 1C,D and Fig. S1D, E, of the preprint). We show that compared to other methods, T-ChIC is able to better preserve the expected biological relationship between the histone modifications and gene expression in single cells.
*In current study, T-ChIC profiled H3K27me3 and H3K4me1 modifications, these data look great. How about other histone modifications (eg H3K9me3 and H3K36me3) and transcription factors? *
While we haven't profiled these other modifications using T-ChIC in Zebrafish, we have previously published high quality data on these histone modifications using the sortChIC method, on which T-ChIC is based (Zeller, Yeung et al 2023). In our comparison, we find that histone modification profiles between T-ChIC and sortChIC are very similar (Fig. S1C in Zeller, Blotenburg et al 2024). Therefore the method is expected to work as well for the other histone marks.
*T-ChIC can detect full length transcription from the same single cells, but in FigS3, the authors still used other published single cell transcriptomics to annotate the cell types, this seems unnecessary? *
We used the published scRNA-seq dataset with a larger number of cells to homogenize our cell type labels with these datasets, but we also cross-referenced our cluster-specific marker genes with ZFIN and homogenized the cell type labels with ZFIN ontology. This way our annotation is in line with previous datasets but not biased by it. Due the relatively smaller size of our data, we didn't expect to identify unique, rare cell types, but our full-length total RNA assay helps us identify non-coding RNAs such as miRNA previously undetected in scRNA assays, which we have now highlighted in new figure S1c .
*Throughout the manuscript, the authors found some interesting dynamics between chromatin state and gene expression during embryogenesis, independent approaches should be used to validate these findings, such as IHC staining or RNA ISH? *
We appreciate that the ISH staining could be useful to validate the expression pattern of genes identified in this study. But to validate the relationships between the histone marks and gene expression, we need to combine these stainings with functional genomics experiments, such as PRC2-related knockouts. Due to their complexity, such experiments are beyond the scope of this manuscript (see also reply to reviewer #3, comment #4 for details).
*In Fig2 and FigS4, the authors showed H3K27me3 cis spreading during development, this looks really interesting. Is this zebrafish specific? H3K27me3 ChIP-seq or CutTag data from mouse and/or human embryos should be reanalyzed and used to compare. The authors could speculate some possible mechanisms to explain this spreading pattern? *
Thanks for the suggestion. In this revision, we have reanalysed a dataset of mouse ChIP-seq of H3K27me3 during mouse embryonic development by Xiang et al (Nature Genetics 2019) and find similar evidence of spreading of H3K27me3 signal from their pre-marked promoter regions at E5.5 epiblast upon differentiation (new Figure S4i). This observation, combined with the fact that the mechanism of pre-marking of promoters by PRC1-PRC2 interaction seems to be conserved between the two species (see (Hickey et al., 2022), (Mei et al., 2021) & (Chen et al., 2021)), suggests that the dynamics of H3K27me3 pattern establishment is conserved across vertebrates. But we think a high-resolution profiling via a method like T-ChIC would be more useful to demonstrate the dynamics of signal spreading during mouse embryonic development in the future. We have discussed this further in our revised manuscript.
Reviewer #1 (Significance (Required)):
*The authors have a longstanding focus and reputation on single cell sequencing technology development and application. In this current study, the authors developed a novel single-cell multi-omic assay termed "T-ChIC" so that to jointly profile the histone modifications along with the full-length transcriptome from the same single cells, analyzed the dynamic relationship between chromatin state and gene expression during zebrafish development and cell fate determination. In general, the assay works well, the data look convincing and conclusions are beneficial to the community. *
Thank you very much for your supportive remarks.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
*Joint analysis of multiple modalities in single cells will provide a comprehensive view of cell fate states. In this manuscript, Bhardwaj et al developed a single-cell multi-omics assay, T-ChIC, to simultaneously capture histone modifications and full-length transcriptome and applied the method on early embryos of zebrafish. The authors observed a decoupled relationship between the chromatin modifications and gene expression at early developmental stages. The correlation becomes stronger as development proceeds, as genes are silenced by the cis-spreading of the repressive marker H3k27me3. Overall, the work is well performed, and the results are meaningful and interesting to readers in the epigenomic and embryonic development fields. There are some concerns before the manuscript is considered for publication. *
We thank the reviewer for appreciating the quality of our study.
*Major concerns: *
- A major point of this study is to understand embryo development, especially gastrulation, with the power of scMulti-Omics assay. However, the current analysis didn't focus on deciphering the biology of gastrulation, i.e., lineage-specific pioneer factors that help to reform the chromatin landscape. The majority of the data analysis is based on the temporal dimension, but not the cell-type-specific dimension, which reduces the value of the single-cell assay. *
We focused on the lineage-specific transcription factor activity during gastrulation in Figure 4 and S8 of the manuscript and discovered several interesting regulators active at this stage. During our analysis of the temporal dimension for the rest of the manuscript, we also classified the cells by their germ layer and "latent" developmental time by taking the full advantage of the single-cell nature of our data. Additionally, we have now added the cell-type-specific H3K27-demethylation results for 24hpf in response to your comment below. We hope that these results, together with our openly available dataset would demonstrate the advantage of the single-cell aspect of our dataset.
- *The cis-spreading of H3K27me3 with developmental time is interesting. Considering H3k27me3 could mark bivalent regions, especially in pluripotent cells, there must be some regions that have lost H3k27me3 signals during development. Therefore, it's confusing that the authors didn't find these regions (30% spreading, 70% stable). The authors should explain and discuss this issue. *
Indeed we see that ~30% of the bins enriched in the pluripotent stage spread, while 70% do not seem to spread. In line with earlier observations(Hickey et al., 2022; Vastenhouw et al., 2010), we find that H3K27me3 is almost absent in the zygote and is still being accumulated until 24hpf and beyond. Therefore the majority of the sites in the genome still seem to be in the process of gaining H3K27me3 until 24hpf, explaining why we see mostly "spreading" and "stable" states. Considering most of these sites are at promoters and show signs of bivalency, we think that these sites are marked for activation or silencing at later stages. We have discussed this in the manuscript ("discussion"). However, in response to this and earlier comment, we went back and searched for genes that show H3K27-demethylation in the most mature cell types (at 24 hpf) in our data, and found a subset of genes that show K27 demethylation after acquiring them earlier. Interestingly, most of the top genes in this list are well-known as developmentally important for their corresponding cell types. We have added this new result and discussed it further in the manuscript (Fig. 2d,e, , Supplementary table 3).
*Minors: *
- The authors cited two scMulti-omics studies in the introduction, but there have been lots of single-cell multi-omics studies published recently. The authors should cite and consider them. *
We have cited more single-cell chromatin and multiome studies focussed on early embryogenesis in the introduction now.
*2. T-ChIC seems to have been presented in a previous paper (ref 15). Therefore, Fig. 1a is unnecessary to show. *
Figure 1a. shows a summary of our Zebrafish TChIC workflow, which contains the unique sample multiplexing and sorting strategy to reduce batch effects, which was not applied in the original TChIC workflow. We have now clarified this in "Results".
- *It's better to show the percentage of cell numbers (30% vs 70%) for each heatmap in Figure 2C. *
We have added the numbers to the corresponding legends.
- *Please double-check the citation of Fig. S4C, which may not relate to the conclusion of signal differences between lineages. *
The citation seems to be correct (Fig. S4C supplements Fig. 2C, but shows mesodermal lineage cells) but the description of the legend was a bit misleading. We have clarified this now.
*5. Figure 4C has not been cited or mentioned in the main text. Please check. *
Thanks for pointing it out. We have cited it in Results now.
Reviewer #2 (Significance (Required)):
*Strengths: This work utilized a new single-cell multi-omics method and generated abundant epigenomics and transcriptomics datasets for cells covering multiple key developmental stages of zebrafish. *
*Limitations: The data analysis was superficial and mainly focused on the correspondence between the two modalities. The discussion of developmental biology was limited. *
*Advance: The zebrafish single-cell datasets are valuable. The T-ChIC method is new and interesting. *
*The audience will be specialized and from basic research fields, such as developmental biology, epigenomics, bioinformatics, etc. *
*I'm more specialized in the direction of single-cell epigenomics, gene regulation, 3D genomics, etc. *
Thank you for your remarks.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
*This manuscript introduces T‑ChIC, a single‑cell multi‑omics workflow that jointly profiles full‑length transcripts and histone modifications (H3K27me3 and H3K4me1) and applies it to early zebrafish embryos (4-24 hpf). The study convincingly demonstrates that chromatin-transcription coupling strengthens during gastrulation and somitogenesis, that promoter‑anchored H3K27me3 spreads in cis to enforce developmental gene silencing, and that integrating TF chromatin status with expression can predict lineage‑specific activators and repressors. *
*Major concerns *
- *Independent biological replicates are absent, so the authors should process at least one additional clutch of embryos for key stages (e.g., 6 hpf and 12 hpf) with T‑ChIC and demonstrate that the resulting data match the current dataset. *
Thanks for pointing this out. We had, in fact, performed T-ChIC experiments in four rounds of biological replicates (independent clutch of embryos) and merged the data to create our resource. Although not all timepoints were profiled in each replicate, two timepoints (10 and 24hpf) are present in all four, and the celltype composition of these replicates from these 2 timepoints are very similar. We have added new plots in figure S2f and added (new) supplementary table (#1) to highlight the presence of biological replicates.
2. *The TF‑activity regression model uses an arbitrary R² {greater than or equal to} 0.6 threshold; cross‑validated R² distributions, permutation‑based FDR control, and effect‑size confidence intervals are needed to justify this cut‑off. *
Thank you for this suggestion. We did use 10-fold cross validation during training and obtained the R2 values of TF motifs from the independent test set as an unbiased estimate. However, the cutoff of R2 > 0.6 to select the TFs for classification was indeed arbitrary. In the revised version, we now report the FDR-adjusted p-values for these R2 estimates based on permutation tests, and select TFs with a cutoff of padj supplementary table #4 to include the p-values for all tested TFs. However, we see that our arbitrary cutoff of 0.6 was in fact, too stringent, and we can classify many more TFs based on the FDR cutoffs. We also updated our reported numbers in Fig. 4c to reflect this. Moreover, supplementary table #4 contains the complete list of TFs used in the analysis to allow others to choose their own cutoff.
3. *Predicted TF functions lack empirical support, making it essential to test representative activators (e.g., Tbx16) and repressors (e.g., Zbtb16a) via CRISPRi or morpholino knock‑down and to measure target‑gene expression and H3K4me1 changes. *
We agree that independent validation of the functions of our predicted TFs on target gene activity would be important. During this revision, we analysed recently published scRNA-seq data of Saunders et al. (2023) (Saunders et al., 2023), which includes CRISPR-mediated F0 knockouts of a couple of our predicted TFs, but the scRNAseq was performed at later stages (24hpf onward) compared to our H3K4me1 analysis (which was 4-12 hpf). Therefore, we saw off-target genes being affected in lineages where these TFs are clearly not expressed (attached Fig 1). We therefore didn't include these results in the manuscript. In future, we aim to systematically test the TFs predicted in our study with CRISPRi or similar experiments.
4. *The study does not prove that H3K27me3 spreading causes silencing; embryos treated with an Ezh2 inhibitor or prc2 mutants should be re‑profiled by T‑ChIC to show loss of spreading along with gene re‑expression. *
We appreciate the suggestion that indeed PRC2-disruption followed by T-ChIC or other forms of validation would be needed to confirm whether the H3K27me3 spreading is indeed causally linked to the silencing of the identified target genes. But performing this validation is complicated because of multiple reasons: 1) due to the EZH2 contribution from maternal RNA and the contradicting effects of various EZH2 zygotic mutations (depending on where the mutation occurs), the only properly validated PRC2-related mutant seems to be the maternal-zygotic mutant MZezh2, which requires germ cell transplantation (see Rougeot et al. 2019 (Rougeot et al., 2019)) , and San et al. 2019 (San et al., 2019) for details). The use of inhibitors have been described in other studies (den Broeder et al., 2020; Huang et al., 2021), but they do not show a validation of the H3K27me3 loss or a similar phenotype as the MZezh2 mutants, and can present unwanted side effects and toxicity at a high dose, affecting gene expression results. Moreover, in an attempt to validate, we performed our own trials with the EZH2 inhibitor (GSK123) and saw that this time window might be too short to see the effect within 24hpf (attached Fig. 2). Therefore, this validation is a more complex endeavor beyond the scope of this study. Nevertheless, our further analysis of H3K27me3 de-methylation on developmentally important genes (new Fig. 2e-f, Sup. table 3) adds more confidence that the polycomb repression plays an important role, and provides enough ground for future follow up studies.
*Minor concerns *
- *Repressive chromatin coverage is limited, so profiling an additional silencing mark such as H3K9me3 or DNA methylation would clarify cooperation with H3K27me3 during development. *
We agree that H3K27me3 alone would not be sufficient to fully understand the repressive chromatin state. Extension to other chromatin marks and DNA methylation would be the focus of our follow up works.
*2. Computational transparency is incomplete; a supplementary table listing all trimming, mapping, and peak‑calling parameters (cutadapt, STAR/hisat2, MACS2, histoneHMM, etc.) should be provided. *
As mentioned in the manuscript, we provide an open-source pre-processing pipeline "scChICflow" to perform all these steps (github.com/bhardwaj-lab/scChICflow). We have now also provided the configuration files on our zenodo repository (see below), which can simply be plugged into this pipeline together with the fastq files from GEO to obtain the processed dataset that we describe in the manuscript. Additionally, we have also clarified the peak calling and post-processing steps in the manuscript now.
*3. Data‑ and code‑availability statements lack detail; the exact GEO accession release date, loom‑file contents, and a DOI‑tagged Zenodo archive of analysis scripts should be added. *
We have now publicly released the .h5ad files with raw counts, normalized counts, and complete gene and cell-level metadata, along with signal tracks (bigwigs) and peaks on GEO. Additionally, we now also released the source datasets and notebooks (.Rmarkdown format) on Zenodo that can be used to replicate the figures in the manuscript, and updated our statements on "Data and code availability".
*4. Minor editorial issues remain, such as replacing "critical" with "crucial" in the Abstract, adding software version numbers to figure legends, and correcting the SAMtools reference. *
Thank you for spotting them. We have fixed these issues.
Reviewer #3 (Significance (Required)):
The method is technically innovative and the biological insights are valuable; however, several issues-mainly concerning experimental design, statistical rigor, and functional validation-must be addressed to solidify the conclusions.
Thank you for your comments. We hope to have addressed your concerns in this revised version of our manuscript.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript introduces T‑ChIC, a single‑cell multi‑omics workflow that jointly profiles full‑length transcripts and histone modifications (H3K27me3 and H3K4me1) and applies it to early zebrafish embryos (4-24 hpf). The study convincingly demonstrates that chromatin-transcription coupling strengthens during gastrulation and somitogenesis, that promoter‑anchored H3K27me3 spreads in cis to enforce developmental gene silencing, and that integrating TF chromatin status with expression can predict lineage‑specific activators and repressors.
Major concerns
Minor concerns
The method is technically innovative and the biological insights are valuable; however, several issues-mainly concerning experimental design, statistical rigor, and functional validation-must be addressed to solidify the conclusions.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Joint analysis of multiple modalities in single cells will provide a comprehensive view of cell fate states. In this manuscript, Bhardwaj et al developed a single-cell multi-omics assay, T-ChIC, to simultaneously capture histone modifications and full-length transcriptome and applied the method on early embryos of zebrafish. The authors observed a decoupled relationship between the chromatin modifications and gene expression at early developmental stages. The correlation becomes stronger as development proceeds, as genes are silenced by the cis-spreading of the repressive marker H3k27me3. Overall, the work is well performed, and the results are meaningful and interesting to readers in the epigenomic and embryonic development fields. There are some concerns before the manuscript is considered for publication.
Major concerns:
Minors:
Strengths: This work utilized a new single-cell multi-omics method and generated abundant epigenomics and transcriptomics datasets for cells covering multiple key developmental stages of zebrafish. Limitations: The data analysis was superficial and mainly focused on the correspondence between the two modalities. The discussion of developmental biology was limited.
Advance: The zebrafish single-cell datasets are valuable. The T-ChIC method is new and interesting.
The audience will be specialized and from basic research fields, such as developmental biology, epigenomics, bioinformatics, etc.
I'm more specialized in the direction of single-cell epigenomics, gene regulation, 3D genomics, etc.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The authors have a longstanding focus and reputation on single cell sequencing technology development and application. In this current study, the authors developed a novel single-cell multi-omic assay termed "T-ChIC" so that to jointly profile the histone modifications along with the full-length transcriptome from the same single cells, analyzed the dynamic relationship between chromatin state and gene expression during zebrafish development and cell fate determination. In general, the assay works well, the data look convincing and conclusions are beneficial to the community.
There are several single-cell methodologies all claim to co-profile chromatin modifications and gene expression from the same individual cell, such as CoTECH, Paired-tag and others. Although T-ChIC employs pA-Mnase and IVT to obtain these modalities from single cells which are different, could the author provide some direct comparisons among all these technologies to see whether T-ChIC outperforms?
In current study, T-ChIC profiled H3K27me3 and H3K4me1 modifications, these data look great. How about other histone modifications (eg H3K9me3 and H3K36me3) and transcription factors?
T-ChIC can detect full length transcription from the same single cells, but in FigS3, the authors still used other published single cell transcriptomics to annotate the cell types, this seems unnecessary?
Throughout the manuscript, the authors found some interesting dynamics between chromatin state and gene expression during embryogenesis, independent approaches should be used to validate these findings, such as IHC staining or RNA ISH?
In Fig2 and FigS4, the authors showed H3K27me3 cis spreading during development, this looks really interesting. Is this zebrafish specific? H3K27me3 ChIP-seq or CutTag data from mouse and/or human embryos should be reanalyzed and used to compare. The authors could speculate some possible mechanisms to explain this spreading pattern?
The authors have a longstanding focus and reputation on single cell sequencing technology development and application. In this current study, the authors developed a novel single-cell multi-omic assay termed "T-ChIC" so that to jointly profile the histone modifications along with the full-length transcriptome from the same single cells, analyzed the dynamic relationship between chromatin state and gene expression during zebrafish development and cell fate determination. In general, the assay works well, the data look convincing and conclusions are beneficial to the community.
Download the complete Review Process [PDF] including:
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Manuscript number: RC-2025-02879 Corresponding author(s): Matteo Allegretti; Alia dos Santos
In this study, we investigated the effects of paclitaxel on both healthy and cancerous cells, focusing on alterations in nuclear architecture. Our novel findings show that:
Paclitaxel-induced microtubule reorganisation during interphase alters the perinuclear distribution of actin and vimentin. The formation of extensive microtubule bundles, in paclitaxel or following GFP-Tau overexpression, coincides with nuclear shape deformation, loss of regulation of nuclear envelope spacing, and alteration of the nuclear lamina.
Paclitaxel treatment reduces Lamin A/C protein levels via a SUN2-dependent mechanism. SUN2, which links the lamina to the cytoskeleton, undergoes ubiquitination and consequent degradation following paclitaxel exposure.
Lamin A/C expression, frequently dysregulated in cancer cells, is a key determinant of cellular sensitivity to, and recovery from, paclitaxel treatment.
Collectively, our data support a model in which paclitaxel disrupts nuclear architecture through two mechanisms: (i) aberrant nuclear-cytoskeletal coupling during interphase, and (ii) multimicronucleation following defective mitotic exit. This represents an additional mode of action for paclitaxel beyond its well-established mechanism of mitotic arrest.
We thank the reviewers for their time and constructive feedback. We have carefully considered all comments and have carried out a full revision. The updated manuscript now includes additional data showing:
Overexpression of microtubule-associated protein Tau causes similar nuclear aberration phenotypes to paclitaxel. This supports our hypothesis that increased microtubule bundling directly leads to nuclear disruption in paclitaxel during interphase.
Paclitaxel's effects on nuclear shape and Lamin A/C and SUN2 expression levels occur independently of cell division.
Reduced levels of Lamin A/C and SUN2 upon paclitaxel treatment occur at the protein level via ubiquitination of SUN2.
The effects of paclitaxel on the nucleus are conserved in breast cancer cells.
Full Revision
We have also edited our text and added further detail to clarify points raised by the reviewers. We believe that our revised manuscript is overall more complete, solid and compelling thanks to the reviewers' comments.
Reviewer #1 Evidence, reproducibility and clarity
This description of the down-regulation of the expression of lamin A/C upon treatment with paclitaxel and its sensitivity to SUN2 is quite interesting but still somehow preliminary. It is unclear whether this effect involves the regulation of gene expression, or of the stability of the proteins. How SUN2 mediates this effect is still unknown.
We thank the reviewer for this valuable comment. To elucidate the mechanism behind the decrease in Lamin A/C and SUN2 levels, we have now performed several additional experiments. First, we performed RT-qPCR to quantify mRNA levels of these genes, relative to the housekeeping gene GAPDH (Supplementary Figure 3B and O). The levels of SUN2 and LMNA mRNA remained the same between control and paclitaxel-treated cells, indicating that this effect instead occurs at the protein level. We have also tested post-translational modifications as a potential regulatory mechanism for Lamin A/C and SUN2. In addition to the phosphorylation of Ser404 which we had already tested (Supplementary Figure 3C), we have now included additional Phos-tag gel and Western blotting data showing that the overall phosphorylation status of Lamin A/C is not affected by paclitaxel (Supplementary Figure 3E and F). We also pulled-down Lamin A/C from cell lysates and then Western blotted for polyubiquitin and acetyl-lysine, which showed that the ubiquitination and acetylation states of Lamin A/C are also not affected by paclitaxel (Supplementary Figure 3G-I). However, Western blots for polyubiquitin of SUN2 pulled down from cell lysates showed that paclitaxel treatment results in significant SUN2 ubiquitination (Figure 3M and N). Therefore, we propose that the downregulation of SUN2 following paclitaxel treatment occurs by ubiquitin-mediated proteolysis.
The roles of free tubulins and polymerized microtubules, and thus the potential role of paclitaxel, need to be uncovered.
We addressed this important point by using an alternative method to stabilise/bundle microtubules in interphase, namely by overexpressing GFP-Tau, as suggested by reviewer 2. Following GFP- Tau overexpression, large microtubule bundles were observed throughout the cytoplasm (Figure 4A), and this resulted in a significant decrease in nuclear solidity (Figure 4B). Furthermore, in cells where microtubule bundles extensively contacted the nucleus, the nuclear lamina became unevenly distributed and appeared patchy (Figure 4C). This supports our hypothesis that the aberrations to nuclear shape and Lamin A/C localisation in paclitaxel-treated cells are due to the presence of microtubules bundles surrounding the nucleus.
The doses of paclitaxel at which occur the effects described in the paper are not fully consistent with all the conclusions. Most experiments have been done at 5 nM. However, at this dose the effect of lamin A/C over or down expression on the growth (differences in the slopes of the curves in Figure 4A) are not fully convincing and not fully consistent with the clear effect on viability as well (in addition, duration of treatments before assessing vialbility are not specified). At 1 nM, cell growth is reduced and the rescuing effect of lamin over-expression is much more clear (Fig 4A), and the nucleus deformation clear (Fig 2A) but this dose has no effect on lamin A/C expression (Fig 3C), which questions how lamins impact nucleus shape and cell survival. Cytoskeleton reorganisation in these conditions is not described although it could clarify the respective role of force production (suggested in figure 1) and nuclei resistance (shown in figure 2) in paclitaxel sensitivity.
We thank the reviewer for raising this important point. We have addressed this by conducting additional repeats for the cell confluency measurements to increase the statistical power of our experiments (Figure 5A). Our data now show that GFP-lamin A/C had a statistically significant effect on rescuing cell growth at both 1 nM and 5 nM paclitaxel, while Lamin A/C knockdown exacerbated the inhibition of cell growth at 5 nM paclitaxel but not 1 nM paclitaxel (Figure 5A). In addition, we note that the duration of paclitaxel treatment before assessing viability was specified in the figure legend: "Bar graph comparing cell viability between wild-type (red), GFP-Lamin A/C overexpression (green), and Lamin A/C knockdown (blue) cells following 20 h incubation in 0, 1, 5, or 10 nM paclitaxel." We also repeated cell viability analysis after 48 h incubation in paclitaxel instead of 20 h to allow for a longer time for differences to take effect (Figure 5B).
We also added figures showing the cytoskeletal reorganisation at both 1 and 10 nM in addition to 0 and 5 nM (Supplementary Figure 1A) showing that microtubule bundling and condensation of actin into puncta correlated with increased paclitaxel concentration. Vimentin colocalised well with microtubules at all concentrations.
We have also included in our results section further clarification for the use of 5nM paclitaxel in this study. The new section reads as follows: "Experiments were performed at 5 nM paclitaxel (with additional experiments to determine dose relationships at 1 and 10 nM) because this aligns with previous studies7,14,24. Furthermore, previous analysis of patient plasma reveals that typical concentrations are within the low nanomolar range8, and concentrations of 5-10 nM are required in cell culture to reach the same intracellular concentrations observed in vivo in patient tumours9. This aligns with in vitro cytotoxic studies of paclitaxel in eight human tumour cell lines which show that paclitaxel's IC50 ranges between 2.5 and 7.5 nM41."
Finally, although the absence of role of mitotic arrest is clear from the data, the defective reorganisation of the nucleus after mitosis still suggest that the effect of paclitaxel is not independent of mitosis.
We thank the reviewer for pointing out the need for clarification in the wording of our manuscript. We have reworded the title and relevant sections of our abstract, introduction, and discussion to make it clearer that the effects of paclitaxel on the nucleus are due to a combination of aberrant nuclear cytoskeletal coupling during interphase and multimicronucleation following mitotic slippage. We have also added additional data in support of the effect of paclitaxel on nuclear architecture during interphase. For this, we used serum-starved cells (which divide only very slowly such that the majority of cells do not pass through mitosis during the 16 h incubation in paclitaxel [Supplementary Figure 2D]). Our new data confirmed that paclitaxel's effects on nuclear solidity, and Lamin A/C and SUN2 proteins levels can occur independently of cell division (Figure 2C; Figure 3H-J). Finally, when we overexpressed GFP-Tau (as discussed above) we observed similar aberrations to nuclear solidity and Lamin A/C localisation. This indicates that these effects occur due to microtubule bundling in interphase, especially as in our study GFP-Tau did not lead to multimicronucleation or appear to affect mitosis (Figure 4).
Below are the main changes to the text regarding the interphase effect of paclitaxel:
Title: "Paclitaxel compromises nuclear integrity in interphase through SUN2-mediated cytoskeletal coupling"
Abstract: "Overall, our data supports nuclear architecture disruption, caused by both aberrant nuclear-cytoskeletal coupling during interphase and exit from defective mitosis, as an additional mechanism for paclitaxel beyond mitotic arrest."
Introduction: "Here we propose that cancer cells have increased vulnerability to paclitaxel both during interphase and following aberrant mitosis due to pre-existing defects in their NE and nuclear lamina."
Discussion: "Overall, our work builds on previous studies investigating loss of nuclear integrity as an anti-cancer mechanism of paclitaxel separate from mitotic arrest14,20,21. We propose that cancer cells show increased sensitivity to nuclear deformation induced by aberrant nuclear-cytoskeletal coupling and multimicronucleation following mitotic slippage. Therefore, we conclude that paclitaxel functions in interphase as well as mitosis, elucidating how slowly growing tumours are targeted."
minor: a more thorough introduction of known data about dose response of cells in culture and in vivo would help understanding the range of concentrations used in this study.
As mentioned above, we have now included additional information in our Results section to clarify our paclitaxel dose range: "Experiments were performed at 5 nM paclitaxel (with additional experiments to determine dose relationships at 1 and 10 nM) because this aligns with previous studies7,14,24. Furthermore, previous analysis of patient plasma reveals that typical concentrations are within the low nanomolar range8, and concentrations of 5-10 nM are required in cell culture to reach the same intracellular concentrations observed in vivo in patient tumours9. This aligns with in vitro cytotoxic studies of paclitaxel in eight human tumour cell lines which show that paclitaxel's IC50 ranges between 2.5 and 7.5 nM41."
Significance
In this manuscript, Hale and colleagues describe the effect of paclitaxel on nucleus deformation and cell survival. They showed that 5nM of paclitaxel induces nucleus fragmentation, cytoskeleton reorganisation, reduced expression of LaminA/C and SUN2, and reduced cell growth and viability. They also showed that these effects could be at least partly compensated by the over-expression of lamin A/C. As fairly acknowledged by the authors, the induction of nuclear deformation in paclitaxel-treated cells, and the increased sensitivity to paclitaxel of cells expressing low level of lamin A/C are not novel (reference #14). Here the authors provided more details on the cytoskeleton changes and nuclear membrane deformation upon paclitaxel treatment. The effect of lamin A/C over and down expression on cell growth and survival are not fully convincing, as further discussed below. The most novel part is the observation that paclitaxel can induce the down-regulation of the expression of lamin A/C and that this effect is mediated by SUN2.
We appreciate the reviewer's summary and thank them for their time. We believe our comprehensive revisions have addressed all comments, strengthening the manuscript and making it more robust and compelling.
Reviewer #2 Evidence, reproducibility and clarity This study investigates the effects of the chemotherapeutic drug paclitaxel on nuclear-cytoskeletal coupling during interphase, claiming a novel mechanism for its anti-cancer activity. The study uses hTERT-immortalized human fibroblasts. After paclitaxel exposure, a suite of state- of-the-art imaging modalities visualizes changes in the cytoskeleton and nuclear architecture. These include STORM imaging and a large number of FIB-SEM tomograms.
We thank the reviewer for the summary and for highlighting our efforts in using the latest imaging technical advances.
Major comments:
The authors make a major claim that in addition to the somewhat well-described mechanism of paclitaxel on mitosis, they have discovered 'an alternative, poorly characterised mechanism in interphase'.
However, none of the data proves that the effects shown are independent of mitosis. To the contrary, measurements are presented 48 hours after paclitaxel treatment starts, after which it can be assumed that 100% of cells have completed at least one mitotic event. The appearance of micronuclei evidences this, as discussed by the authors shortly. It looks like most of the results shown are based on botched mitosis or, more specifically, errors on nuclear assembly upon exit from mitosis rather than a specific effect of paclitaxel on interphase. The readouts the authors show just happen to be measurements while the cells are in interphase.
Alternative hypotheses are missing throughout the manuscript, and so are critical controls and interpretations.
We thank the reviewer for highlighting the lack of clarity in our wording. We have revised the title, abstract and relevant sections of the introduction and discussion to clarify our message that the effects of paclitaxel on the nucleus arise from a combination of aberrant nuclear-cytoskeletal coupling during interphase and multimicronucleation following exit from defective mitosis. We have also included additional data where we used slow-dividing, serum-starved cells (under these conditions, the majority of cells do not undergo mitosis during the 16 h incubation in paclitaxel [Supplementary Figure 2D]). Our new data show that even in these cells there is a clear effect of paclitaxel on nuclear solidity, and Lamin A/C and SUN2 protein levels, further supporting our hypothesis that these phenotypes can occur independently of cell division (Figure 2C; Figure 3H-J). Furthermore, we performed additional experiments where we used overexpression of GFP-Tau as an alternative method of stabilising microtubules in interphase and observed similar aberrations to nuclear solidity and Lamin A/C localisation. As GFP-Tau overexpression did not lead to micronucleation or appear to affect mitosis, these data support the hypothesis that nuclear aberrations occur due to microtubule bundling in interphase (Figure 4). We discuss these experiments in more detail below. Finally, we have reworded the introduction to better introduce alternative hypotheses and mechanisms for paclitaxel's activity.
The authors claim that 'Previously, the anti-cancer activity of paclitaxel was thought to rely mostly on the activation of the mitotic checkpoint through disruption of microtubule dynamics, ultimately resulting in apoptosis.' The authors may have overlooked much of the existing literature on the topic, including many recent manuscripts from Xiang-Xi Xu's and another lab.
We would like to note that the paper from Xiang-Xi Xu's lab (Smith et al, 2021) was cited in our original manuscript (reference 14 in both the original and revised manuscripts). We have now also included additional review articles from the Xiang-Xi Xu lab (PMID:36368286 20 and PMID: 35048083 21). Furthermore, we have clarified the wording in both the introduction and discussion to better reflect the current understanding of paclitaxel's mechanism and alternative hypotheses.
The data, e.g. in Figure 1, does not hold up to the first alternative hypothesis, e.g. that paclitaxel stabilizes microtubules and that excessive mechanical bundling of microtubules induces major changes to cell shape and mechanical stress on the nucleus. Even the simplest controls for this effect (the application of an alternative MT stabilizing drug or the overexpression of an MT stabilizer, e.g., tau).
We thank the reviewer for suggesting this control experiment using the microtubule stabiliser Tau. We have now included these experiments in the revised version of the manuscript (Figure 4). The overexpression of GFP-Tau supports our hypothesis that cytoskeletal reorganisation in paclitaxel exerts mechanical stress on the nucleus during interphase, resulting in nuclear deformation and aberrations to the nuclear lamina. In particular, GFP-Tau overexpression resulted in large microtubule bundles throughout the cytoplasm (Figure 4A). Notably, in cells where these bundles extensively contacted the nucleus, we observed a significant decrease in nuclear solidity (Figure 4B) accompanied by changes in nuclear lamina organisation, including a patchy lamina phenotype, similar to that induced by paclitaxel (Figure 4C).
The focus on nuclear lamina seems somewhat arbitrary and adjacent to previously published work by other groups. What would happen if the authors stained for focal adhesion markers? There would probably be a major change in number and distribution. Would the authors conclude that paclitaxel exerts a specific effect on focal adhesions? Or would the conclusion be that microtubule stabilization and the following mechanical disruption induce pleiotropic effects in cells? Which effects are significant for paclitaxel function on cancer cells?
We thank the reviewer for raising important points regarding the specificity of paclitaxel's effects. We agree that microtubule stabilisation can induce myriad cellular changes, including alterations to focal adhesions and other cytoskeletal components. Our focus on Lamin A/C and nuclear morphology is grounded both in the established clinical relevance of nuclear mechanics in cancer and builds on mechanistic work from other groups.
Lamin A/C expression is commonly altered in cancer, and nuclear morphology is frequently used in cancer diagnosis35. Lamin A/C also plays a crucial role in regulating nuclear mechanics32 and, importantly, determines cell sensitivity to paclitaxel14. However, the mechanism by which Lamin A/C determines sensitivity of cancer cells to paclitaxel is unclear.
Our data are consistent with Lamin A/C being a determinant of paclitaxel survival sensitivity. We also provide evidence that paclitaxel itself reduces Lamin A/C protein levels and disrupts its organisation at the nuclear envelope. We directly link these effects to microtubule bundling around the nucleus and degradation of force-sensing LINC component SUN2, highlighting the importance of nuclear architecture and mechanics to overall cellular function. Furthermore, we show that recovery from paclitaxel treatment depends on Lamin A/C expression levels. This has clinical relevance, as unlike cancer cells, healthy tissue with non-aberrant lamina would be able to selectively recover from paclitaxel treatment.
Minor comments:
While I understand the difficulty of the experiments and the effort the authors have put into producing FIB-SEM tomograms, I am not sure they are helping their study or adding anything beyond the light microscopy images. Some of the images may even be in the way, such as supplementary Figure 6, which lacks in quality, controls, and interpretation. Do I see a lot of mitochondria in that slice?
We agree with the reviewer that Supplementary Figure 6 does not add significant value to the manuscript and thank the reviewer for pointing this out. We have removed it from the manuscript accordingly.
I may have overlooked it, but has the number of cells from which lamellae have been produced been stated?
We thank the reviewer for pointing out the missing information. For our cryo-ET experiments, we collected data from 9 lamellae from paclitaxel-treated cells and 6 lamellae from control cells, with each lamella derived from a single cell. This information has now been added to the figure legend (Figure 2F).
Significance
The significance of studying the effect of paclitaxel, the most successful chemotherapy drug, should be broad and of interest to basic researchers and clinicians.
As outlined above, I believe that major concerns about the design and interpretation of the study hamper its significance and advancements.
We appreciate the reviewer's concerns and have performed major revisions to strengthen the significance of our study. Specifically, we conducted two key sets of experiments to validate our original conclusions: serum starvation to control for the effects of cell division, and overexpression of the microtubule stabiliser Tau to demonstrate that paclitaxel can affect the nucleus via its microtubule bundling activity in interphase.
By elucidating the mechanistic link between microtubule stabilisation and nuclear-cytoskeletal coupling, our findings contribute to our understanding of paclitaxel's multifaceted actions in cancer cells.
My areas of expertise could be broadly defined as Cell Biology, Cytoskeleton, Microtubules, and Structural Biology.
Reviewer #3 Evidence, reproducibility and clarity The manuscript presents interesting new ideas for the mechanism of an old drug, taxol, which has been studied for the last 40 years.
We thank the reviewer for the positive feedback.
Although similar ideas are published, which may be suitable to be cited? • Paclitaxel resistance related to nuclear envelope structural sturdiness. Smith ER, Wang JQ, Yang DH, Xu XX. Drug Resist Updat. 2022 Dec;65:100881. doi: 10.1016/j.drup.2022.100881. Epub 2022 Oct 15. PMID: 36368286 Review. • Breaking malignant nuclei as a non-mitotic mechanism of taxol/paclitaxel. Smith ER, Xu XX. J Cancer Biol. 2021;2(4):86-93. doi: 10.46439/cancerbiology.2.031. PMID: 35048083 Free PMC article.
We thank the reviewer for bringing to our attention these important review articles. In our initial manuscript, we only cited the original paper (14, also reference 14 in the original manuscript). We have now included citations to the suggested publications (20,21).
We would also like to emphasise how our manuscript distinguishes itself from the work of Smith et al.14,20,21:
Cell-type focus: In their study 14, Smith et al. examined the effect of paclitaxel on malignant ovarian cancer cells and proposed that paclitaxel's effects on the nucleus are limited to cancer cells. However, our data extends these findings by demonstrating paclitaxel's effects in both cancerous and non-cancerous backgrounds.
Cytoskeletal reorganisation: Smith et al. show reorganisation of microtubules in paclitaxel-treated cells14. Our data show re-organisation of other cytoskeletal components, including F-actin and vimentin.
Multimicronucleation: Smith et al. propose that paclitaxel-induced multimicronucleation occurs independently of cell division14. Although we observe progressive nuclear abnormalities during interphase over the course of paclitaxel treatment, our data do not support this conclusion; we find that multimicronucleation occurs only following mitosis.
Direct link between microtubule bundling and nuclear aberrations: We show that nuclear aberrations caused by paclitaxel during interphase (distinct from multimicronucleation) are directly linked to microtubule bundling around the nucleus, suggesting they result from mechanical disruption and altered force propagation.
Lamin A/C regulation: Consistent with Smith et al.14, we show that Lamin A/C depletion leads to increased sensitivity to paclitaxel treatment. However, we further demonstrate that paclitaxel itself leads to reduced levels of Lamin A/C and that this effect occurs independently of mitosis and is mediated via force-sensing LINC component SUN2. Upon SUN2 knockdown, Lamin A/C levels are no longer affected by paclitaxel treatment.
Recovery: Finally, our work reveals that cells expressing low levels of Lamin A/C recover less efficiently after paclitaxel removal. This might help explain how cancer cells could be more susceptible to paclitaxel.
Only one cell line was used in all the experiments? "Human telomerase reverse transcriptase (hTERT) immortalised human fibroblasts" ? The cells used are not very relevant to cancer cells (carcinomas) that are treated with paclitaxel. It is not clear if the observations and conclusions will be able to be generalized to cancer cells.
We thank the reviewer for this comment. Our initial study aimed to understand the effects of paclitaxel on nuclear architecture in non-aberrant backgrounds. To show that the observed effects of paclitaxel are also applicable to cancer cells, we have now repeated our main experiments using MDA-MB-231 human breast cancer cells (Supplementary Figure 1B; Supplementary Figure 3P-T). Similar to our findings in human fibroblasts, paclitaxel treatment of MDA-MB-231 led to cytoskeletal reorganisation (Supplementary Figure 1B), a decrease in nuclear solidity (Supplementary Figure 3P), aberrant (patchy) localisation of Lamin A/C (Supplementary Figure 3Q), and a reduction in Lamin A/C and SUN2 levels (Supplementary Figure 3R-T).
"Fig. 1. (B) STORM imaging of α-tubulin immunofluorescence in cells fixed after 16 h incubation in control media or 5 nM paclitaxel. Lower panels show α-tubulin clusters generated with HDBSCAN analysis. Scale bars = 10 μm." It needs explanation of what is meaning of the different color lines in the lower panels, just different filaments?
We have added further detail to the figure legend for clarification: "Lower panels show α-tubulin clusters generated with HDBSCAN analysis. Different colours distinguish individual α-tubulin clusters, representing individual microtubule filaments or filament bundles."
Generally, the figures need additional description to be clear.
We have added further clarification and detail to our figure legends.
"Figure 3 - Paclitaxel results in aberrations to the nuclear lamina." The sentence seems not to be well constructed. "Paclitaxel treatment causes ..."?
We changed this sentence to: "Figure 3 - Paclitaxel treatment results in aberrant organisation of the nuclear lamina and decreased Lamin A/C levels via SUN2."
Lamin A and C levels are different in different images (Fig. 3B, H): some Lamin A is higher, and sometime Lamin C is higher? This may possibly due to culture condition or subtle difference in sample handling?.
We thank the reviewer for pointing this out and we agree that the ratio of Lamin A to Lamin C can vary with culture conditions. To confirm that paclitaxel treatment reduces total Lamin A/C levels regardless of this ratio, we repeated the Western blot analysis in three additional biological replicates using cells in which Lamin C levels exceeded Lamin A levels. These experiments confirmed a comparable decrease in total Lamin A/C levels. Figure 3B and 3C have been updated accordingly.
Also, the effect on Lamin A/C and SUN2 levels are not significant of robust.
Decreased Lamin A/C and SUN2 levels following paclitaxel treatment were consistently seen across three or more biological repeats (Figure 3B-C), and this could be replicated in a different cell type (MDA-MB-231) (Supplementary Figure 3R-T). Furthermore, Western blotting results are consistent with the patchy Lamin A/C distribution observed using confocal and STORM following paclitaxel treatment (Figure 3A; Supplementary Figure 3A), where Lamin A/C appears to be absent from discrete areas of the lamina.
Any mechanisms are speculated for the reason for the reduction?
We have now included additional data which aims to shed light on the mechanism behind the decrease in Lamin A/C and SUN2 levels following paclitaxel treatment. We found that SUN2 is selectively degraded during paclitaxel treatment. Immunoprecipitation of SUN2 followed by Western blotting against Polyubiquitin C showed increased SUN2 ubiquitination in paclitaxel (Figure 3M and N). Furthermore, in our original manuscript, we showed that Lamina A/C levels remained unaltered during paclitaxel treatment in cells where SUN2 had been knocked down. We propose that changes in microtubule organisation affect force propagation to Lamin A/C specifically via SUN2 and that this leads to Lamina A/C removal and depletion. Future work will be needed to fully understand this mechanism.
In addition to the findings described above, we report no significant changes in mRNA levels for LMNA or SUN2 in paclitaxel (Supplementary Figure 3B and O). Phos-tag gels followed by Western blotting analysis for Lamin A/C also did not detect changes to the overall phosphorylation status of Lamin A/C due to paclitaxel treatment. This is in agreement with our initial data showing no changes to Lamin A/C Ser 404 phosphorylation levels (Supplementary Figure 3E and F). Finally, Lamin A/C immunoprecipitation experiments followed by Western blotting for Polyubiquitin C and acetyl-lysine showed no significant changes in the ubiquitination and acetylation state of Lamin A/C in paclitaxel-treated cells (Supplementary Figure 3G-I).
Also, the about 50% reduction in protein level is difficult to be convincing as an explanation of nuclear disruption.
The nuclear lamina and LINC complex proteins play a critical role in regulating nuclear integrity, stiffness and mechanical responsiveness to external forces28,31-33,54,75, as well as in maintaining the nuclear intermembrane distance69,74. In particular, SUN-domain proteins physically bridge the nuclear lamina to the cytoskeleton through interactions with Nesprins, thereby preserving the perinuclear space distance30,69,74. Mutations in Lamins have been shown to disrupt chromatin organization, alter gene expression, and compromise nuclear structural integrity, and experiments with LMNA knockout cells reveal that nuclear mechanical fragility is closely coupled to nuclear deformation47. Furthermore, nuclear-cytoskeletal coupling is essential during processes such as cell migration, where cells undergo stretching and compression of the nucleus; weakening or loss of the lamina in such cases compromises cell movement47,73. In our work, we show that alterations to nuclear Lamin A/C and SUN2 by paclitaxel treatment coincide with nuclear deformations (Figure 2A-D, F, G; Figure 3A-D, F, G; Supplementary Figure 3A, P-T) and that these deformations are reversible following paclitaxel removal (Supplementary Figure 4B-D). Our experiments also demonstrate that Lamin A/C expression levels significantly influence cell growth, cell viability, and cell recovery in paclitaxel (Figure 5). Therefore, drawing on current literature and our results, we propose that, during interphase, paclitaxel induces severe nuclear aberrations through the combined effects of: i) increased cytoskeletal forces on the NE caused by microtubule bundling; ii) loss of ~50% Lamin A/C and SUN2; iii) reorganisation of nucleo-cytoskeletal components.
Significance
The manuscript presents interesting new ideas for the mechanism of an old drug, taxol, which has been studied for the last 40 years.
The data may be improved to provide stronger support.
Additional cell lines (of cancer or epithelial origin) may be repeated to confirm the generality of the observation and conclusions.?
We thank the reviewer for the feedback and valuable suggestions. In response, we have included experiments using human breast cancer cell line MDA-MB-231 to further corroborate our findings and interpretations. We believe these additions have improved the clarity, robustness and impact of our manuscript, and we are grateful for the reviewer's contributions to its improvement.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript presents interesting new ideas for the mechanism of an old drug, taxol, which has been studied for the last 40 years. Although similar ideas are published, which may be suitable to be cited?
Only one cell line was used in all the experiments? "Human telomerase reverse transcriptase (hTERT) immortalised human fibroblasts" ? The cells used are not very relevant to cancer cells (carcinomas) that are treated with paclitaxel. It is not clear if the observations and conclusions will be able to be generalized to cancer cells.
"Fig. 1. (B) STORM imaging of α-tubulin immunofluorescence in cells fixed after 16 h incubation in control media or 5 nM paclitaxel. Lower panels show α-tubulin clusters generated with HDBSCAN analysis. Scale bars = 10 μm." It needs explanation of what is meaning of the different color lines in the lower panels, just different filaments?
Generally, the figures need additional description to be clear.
"Figure 3 - Paclitaxel results in aberrations to the nuclear lamina." The sentence seems not to be well constructed. "Paclitaxel treatment causes ..."?
Lamin A and C levels are different in different images (Fig. 3B, H): some Lamin A is higher, and sometime Lamin C is higher? This may possibly due to culture condition or subtle difference in sample handling?. Also, the effect on Lamin A/C and SUN2 levels are not significant of robust. Any mechanisms are speculated for the reason for the reduction? Also, the about 50% reduction in protein level is difficult to be convincing as an explanation of nuclear disruption.
The manuscript presents interesting new ideas for the mechanism of an old drug, taxol, which has been studied for the last 40 years.
The data may be improved to provide stronger support.
Additional cell lines (of cancer or epithelial origin) may be repeated to confirm the generality of the observation and conclusions.?
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This study investigates the effects of the chemotherapeutic drug paclitaxel on nuclear-cytoskeletal coupling during interphase, claiming a novel mechanism for its anti-cancer activity. The study uses hTERT-immortalized human fibroblasts. After paclitaxel exposure, a suite of state-of-the-art imaging modalities visualizes changes in the cytoskeleton and nuclear architecture. These include STORM imaging and a large number of FIB-SEM tomograms.
Major comments:
The authors make a major claim that in addition to the somewhat well-described mechanism of paclitaxel on mitosis, they have discovered 'an alternative, poorly characterised mechanism in interphase'.
However, none of the data proves that the effects shown are independent of mitosis. To the contrary, measurements are presented 48 hours after paclitaxel treatment starts, after which it can be assumed that 100% of cells have completed at least one mitotic event. The appearance of micronuclei evidences this, as discussed by the authors shortly. It looks like most of the results shown are based on botched mitosis or, more specifically, errors on nuclear assembly upon exit from mitosis rather than a specific effect of paclitaxel on interphase. The readouts the authors show just happen to be measurements while the cells are in interphase.
Alternative hypotheses are missing throughout the manuscript, and so are critical controls and interpretations.
The authors claim that 'Previously, the anti-cancer activity of paclitaxel was thought to rely mostly on the activation of the mitotic checkpoint through disruption of microtubule dynamics, ultimately resulting in apoptosis.' The authors may have overlooked much of the existing literature on the topic, including many recent manuscripts from Xiang-Xi Xu's and another lab.
The data, e.g. in Figure 1, does not hold up to the first alternative hypothesis, e.g. that paclitaxel stabilizes microtubules and that excessive mechanical bundling of microtubules induces major changes to cell shape and mechanical stress on the nucleus. Even the simplest controls for this effect (the application of an alternative MT stabilizing drug or the overexpression of an MT stabilizer, e.g., tau).
The focus on nuclear lamina seems somewhat arbitrary and adjacent to previously published work by other groups. What would happen if the authors stained for focal adhesion markers? There would probably be a major change in number and distribution. Would the authors conclude that paclitaxel exerts a specific effect on focal adhesions? Or would the conclusion be that microtubule stabilization and the following mechanical disruption induce pleiotropic effects in cells? Which effects are significant for paclitaxel function on cancer cells?
Minor comments:
While I understand the difficulty of the experiments and the effort the authors have put into producing FIB-SEM tomograms, I am not sure they are helping their study or adding anything beyond the light microscopy images. Some of the images may even be in the way, such as supplementary Figure 6, which lacks in quality, controls, and interpretation. Do I see a lot of mitochondria in that slice?
I may have overlooked it, but has the number of cells from which lamellae have been produced been stated?
The significance of studying the effect of paclitaxel, the most successful chemotherapy drug, should be broad and of interest to basic researchers and clinicians.
As outlined above, I believe that major concerns about the design and interpretation of the study hamper its significance and advancements.
My areas of expertise could be broadly defined as Cell Biology, Cytoskeleton, Microtubules, and Structural Biology.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This description of the down-regulation of the expression of lamin A/C upon treatment with paclitaxel and its sensitivity to SUN2 is quite interesting but still somehow preliminary. It is unclear whether this effect involves the regulation of gene expression, or of the stability of the proteins. How SUN2 mediates this effect is still unknown. The roles of free tubulins and polymerized microtubules, and thus the potential role of paclitaxel, need to be uncovered.
The doses of paclitaxel at which occur the effects described in the paper are not fully consistent with all the conclusions. Most experiments have been done at 5 nM. However, at this dose the effect of lamin A/C over or down expression on the growth (differences in the slopes of the curves in Figure 4A) are not fully convincing and not fully consistent with the clear effect on viability as well (in addition, duration of treatments before assessing vialbility are not specified). At 1 nM, cell growth is reduced and the rescuing effect of lamin over-expression is much more clear (Fig 4A), and the nucleus deformation clear (Fig 2A) but this dose has no effect on lamin A/C expression (Fig 3C), which questions how lamins impact nucleus shape and cell survival. Cytoskeleton reorganisation in these conditions is not described although it could clarify the respective role of force production (suggested in figure 1) and nuclei resistance (shown in figure 2) in paclitaxel sensitivity.
Finally, although the absence of role of mitotic arrest is clear from the data, the defective reorganisation of the nucleus after mitosis still suggest that the effect of paclitaxel is not independent of mitosis.
minor: a more thorough introduction of known data about dose response of cells in culture and in vivo would help understanding the range of concentrations used in this study.
In this manuscript, Hale and colleagues describe the effect of paclitaxel on nucleus deformation and cell survival. They showed that 5nM of paclitaxel induces nucleus fragmentation, cytoskeleton reorganisation, reduced expression of LaminA/C and SUN2, and reduced cell growth and viability. They also showed that these effects could be at least partly compensated by the over-expression of lamin A/C. As fairly acknowledged by the authors, the induction of nuclear deformation in paclitaxel-treated cells, and the increased sensitivity to paclitaxel of cells expressing low level of lamin A/C are not novel (reference #14). Here the authors provided more details on the cytoskeleton changes and nuclear membrane deformation upon paclitaxel treatment. The effect of lamin A/C over and down expression on cell growth and survival are not fully convincing, as further discussed below. The most novel part is the observation that paclitaxel can induce the down-regulation of the expression of lamin A/C and that this effect is mediated by SUN2.
Download the complete Review Process [PDF] including:
Download the complete Review Process [PDF] including:
Download the complete Review Process [PDF] including:
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Manuscript number: RC-2025-02946
Corresponding author(s): Margaret, Frame
Roza, Masalmeh
[Please use this template only if the submitted manuscript should be considered by the affiliate journal as a full revision in response to the points raised by the reviewers.
If you wish to submit a preliminary revision with a revision plan, please use our "Revision Plan" template. It is important to use the appropriate template to clearly inform the editors of your intentions.]
This section is optional. Insert here any general statements you wish to make about the goal of the study or about the reviews.
We thank the reviewers for recognizing the significance of our work and for their constructive feedback and suggestions, most of which we have implemented in our revised manuscript.
This section is mandatory. *Please insert a point-by-point reply describing the revisions that were already carried out and included in the transferred manuscript. *
Reviewer #1
Evidence, reproducibility and clarity
Review of Masalmeh et al. Title: "FAK modulates glioblastoma stem cell energetics..."
Previous studies have implicated FAK and the related tyrosine kinase PYK2 in glioblastoma growth, cell migration, and invasion. Herein, using a murine stem cell model of glioblastoma, the authors used CRISPR to inactivate FAK, FAK-null cells selected and cloned, and lentiviral re-expression of murine FAK in the FAK-null cells (termed FAK Rx) was accomplished. FAK-/- cells were shown to possess epithelial characteristics whereas FAK Rx cells expressed mesenchymal markers and increased cell migration/invasion in vitro. Comparisons between FAK-/- and FAK Rx cells showed that FAK re-expressed increased mitochondrial respiration and amino acid uptake. This was associated with FAK Rx cells exhibiting filamentous mitochondrial morphology (potentially an OXPHOS phenotype) and decreased levels of MTFR1L S235 phosphorylation (implicated in mito morphology fragmentation). Mito and epithelial cell morphology of FAK-/- cells was reversed by treatment with Rho-kinase inhibitors that also increased mito metabolism and cell viability. Last, FAK-dependent glioblastoma tumor growth was shown by comparisons of FAK-/- and FAK Rx implantation studies.
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
Some questions that would enhance potential impact. 1. Cell generation. Please describe the analysis of FAK-/- clones in more detail. The "low viability" phenotype needs further explanation with regard to clonal expansion and growth characteristics?
Response:
Figure 1F: need further support of MET change upon FAK KO and EMT reversion.
Response: We have added a heatmap (Figure S1E) illustrating the changes in protein expression of core-enriched EMT/MET genes products (by proteomics) after FAK gene deletion (EMT genes as defined in Howe et al., 2018) ; this strengthens the conclusion that the MET reversion morphological phenotype is accompanied by recognised MET protein changes.
Fig. 2: Need further support if FAK effects impact glycolysis or oxidative phosphorylation in particular as implicated by the stem cell model.
Response: We show that FAK impacts both glycolysis (Figure 2A, 2E, and 2F) and mitochondrial oxidative phosphorylation on the basis of the oxygen consumption rate (OCR) (Figure 2B, and 2D), showing both are contributing pathways to FAK-dependent energy production. We have clarified this in the text.
Is there a combinatorial potential between FAKi and chemotherapies used for glioblastoma. Need to build upon past studies.
Response: Yes, previous studies suggest that inhibiting FAK can sensitize GBM cells to chemotherapy (Golubovskaya et al., 2012; Ortiz-Rivera et al., 2023). We have included a paragraph in the discussion section to make sure this is clearer. Although it is not the subject of this study, we appreciate it is useful context.
The notation of changes in glucose transporter expression should be followed up with regard to the potential that FAK-expressing cells may have different uptake of carbon sources and other amino acids. Altered uptake could be one potential explanation for increase glycolysis and glutamine flux.
Response: We agree with the reviewer that glucose uptake could be contributing and we include data that 2 glucose transporters are indeed FAK-regulated namely Glucose transporter 1 (GLUT1, encoded by Slc2a1 gene) and Glucose transporter 3 (GLUT 3, encoded by Slc2a3 gene) (shown in Figure S2B and C).
It would be helpful to support the confocal microscopy of mitos with EM.
Response:
We are concerned (and in our experience) that Electron microscopy (EM) may introduce artefacts during sample preparation. In contrast, immunofluorescence sample preparation is less susceptible to artefacts. The SORA system we used is not a conventional point-scanning confocal microscope, but is a super-resolution module based on a spinning disk confocal platform (CSU-W1; Yokogawa) using optical pixel reassignment with confocal detection. This method enhances resolution in all dimensions with resolution in our samples measured at 120nm. This has been instructive in defining a new level of changes in mitochondrial morphology upon FAK gene deletion.
Lack of FAK expression with increased MTFR1 phosphorylation is difficult to interpret.
Response: We do not directly show that this phosphorylation event is causal in our experiments; however, we think it important to document this change since it has been published that phosphorylation of MTFR1 has been causally linked to the mitochondrial morphology we observed in other systems (Tilokani et al., 2022).
Need to have better support between loss of FAK and the increase in Rho signaling. Use of Rho kinase inhibitors is very limited and the context to FAK (and or Pyk2) remains unclear. Past studies have linked integrin adhesion to ECM as a linkage between FAK activation and the transient inhibition of RhoA GTP binding. Is integrin signaling and FAK involved in the cell and metabolism phenotypes in this new model?
Response: To better support the antagonistic effect of FAK on Rho-kinase (ROCK) signalling, we included a new experiment in which the integrin-FAK signalling pathway has been disrupted by treating FAK WT cells with an agent that causes detachment from the substratum, Accutase, and growing the cells in suspension in laminin-free medium. We present ROCK activity data, as judged by phosphorylated MLC2 at serine 19 (pMLC2 S19), relating this to induced FAK phosphorylation at Y397 (a surrogate for FAK activity) that is supressed after integrin disengagement. These measurements have been compared with conditions whereby integrin-FAK signalling is activated by growing the cells on laminin coated surfaces. We observed a time-dependent decrease in pFAK(Y397) levels (normalised to total FAK) in suspended cells compared to those spread on laminin, while pMLC2(S19) levels increased in a reciprocal manner over time in detached cells relative to spread cells (S4A and B). There is therefore an inverse relationship between integrin-FAK signalling and ROCK-MLC2 activity, consistent with findings from FAK gene deletion experiments. In the former case, we do not rely on gene deletion cell clones.
Significance
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
__Response: __
Deleting the gene encoding FAK in mouse embryonic fibroblasts leads to elevated Pyk2 expression (Sieg, 2000). However, in the GBM stem cell model we used here, Pyk2 was not expressed (determined by both transcriptomics and proteomics). We have included Figure S1E to show that PYK2 expression was undetectable in FAK -/- and FAK Rx cells at the RNA level (Figure S1F). We conclude that there is no compensatory increase in Pyk2 upon FAK loss in these cells. In the transformed neural stem cell model of GBM, we do not consistently or robustly detect nuclear FAK.
Review #2
Masalmeh and colleagues employ a neural stem/progenitor cell-based glioma model (NPE cells) to investigate the role of Focal Adhesion Kinase (FAK) in GBM, with a focus on potential links between the regulation of morphological/adhesive and metabolic GBM cell properties. For this, the authors employ wt cells alongside newly generated FAK-KO and -reexpressing cells, as well as pharmacological interventions to probe the relevance of specific signaling pathways. The authors´ main claims are that FAK crucially modulates glioma cell morphology, cell-cell and cell-substrate interactions and motility, as well as their metabolism, and that these effects translate to changes to relevant in vivo properties such as invasion and tumor growth.
My main issues are with the model chosen by the authors.
As per the methods section, generation of FAK-KO and -"Rx" NPE cells entailed protracted selection/expansion processes, which may have resulted in inadvertent selection for cellular/molecular properties unrelated to the desired one (loss or gain of FAK expression) and which may have had cascading effects on NPE cells. The authors nonetheless repeatedly claim the parameters they quantify, such as mitochondrial or cytoskeletal properties or metabolic features, to have directly resulted from FAK loss or reintroduction. Examples of such causal inferences are to be found in lines 123, 134/135, 165, 181. Such causal claims are, in my view, unsupported.
Acute perturbation of FAK expression/activity, genetically or pharmacologically, followed by a rapid assessment of the processes under investigation, would be needed to begin to assess causality, even if acute genetic perturbations may be technically challenging as sufficient gene expression reduction or restoration to physiologically relevant levels may be hard to achieve.
Response:
We would like to first comment on the model we used here, which we think will clarify the validity of our approach. The model is a transformed stem cell model of GBM that was published in (Gangoso et al., Cell, 2021) and is now used regularly in the GBM field. As mentioned in the response to Reviewer 1, we have added text (page 4 and 5 in the revised manuscript) and a new supplementary figure (Figure S1D) clarifying that the morphological changes we observed were consistent across multiple FAK -/- clones, showing this was not due to any inter-clonal variability. We also added images showing that the morphological changes were apparent at 48 h after nucleofecting FAK -/- cells with the FAK‑expressing vector specifically (not the empty vector), prior to starting G418 selection to enrich for FAK‑expressing cells (Figure S1C), addressing the worry that clonal variation and selection was the cause of the FAK-dependent phenotypes we observed. We believe that our model provides a type of well controlled, clean genetic cancer cell system of a type that is commonly used in cancer cell biology, allowing us to attribute phenotypes to individual proteins.
We have also carried out a more acute treatment by using the FAK inhibitor VS4718 to perturb FAK kinase activity and assessed the effects on glycolysis and glutamine oxidation after 48h treatment (Figure S2D, E and F). We found that treating the transformed neural stem cells (parental population) with FAK inhibitor (300nM VS4718) decreases glucose incorporation into glycolysis intermediates and glutamine incorporation into TCA cycle intermediates, consistent with a role for FAK's kinase activity in maintaining glycolysis and glutamine oxidation.
The employed pharmacological modulation of ROCK activity is the only approach that, given the presumably acute nature of the treatment, may have allowed the authors to probe the proposed functional links. The methods section of the manuscript does not however comprise details as to the duration of these treatments, which leaves open the possibility of long-term treatment having been carried out (data shown in Figure 5B refers to 72hr treatment).
__Response: __
We have added the duration of the treatment to the Methods section and Figure Legends, to clarify that cells were treated with ROCK inhibitors for 24h, before assessing the effects on mictochondria (Figure 4C, D, S4C and D) and glutamine oxidation (Figure 5A, and S5). For metabolic activity by AlamarBlue assay, cells were treated with ROCK inhibitors for 72h (Figure 5B).
Even in the case of ROCK inhibitor experiments, it is however unclear if and how the effects on cell morphology and adhesion, mitochondrial organization and metabolic activity may be connected to each other and, if at all, to FAK expression.
Given the above uncertainties due to the nature of the model and experimental approaches, it is hard to assess the reliability and thus the relevance of the findings.
Response:
FAK suppresses ROCK activity (as judged by pMLC2 S19, Figure 4A and B). Treating FAK -/- cells with two different ROCK inhibitors restored mesenchymal-like cell morphology, mitochondrial morphology and glutamine oxidation. As mentioned above, to strengthen our evidence for the antagonistic role of FAK in ROCK-MLC2 signalling, we have now introduced an experiment whereby integrin-FAK signalling was disrupted through treatment with a detachment agent (Accutase), and subsequently maintaining the cells in suspension in laminin-free medium. We assessed pMLC2 S19 levels (a measure of ROCK activity) relating this to FAK phosphorylation that is supressed after integrin disengagement. These results were evaluated relative to spread wild type cells growing on laminin where Integrin-FAK signalling was active (Figure S4A and B). We observed an inverse relationship between Integrin-FAK signalling and ROCK-MLC2 activity in keeping with our conclusions (Figure 4A and B).
Experimental support for the ability of cell-substrate interaction modulation to concomitantly impact cellular metabolism and motility/invasion would be significant both in terms of advancing our understanding of glioma cell biology and of its translational potential, but the evidence being provided is at best compatible with the proposed model.
Response: We carried out a new experiment to support the ability of cell-substrate interaction modulation to impact metabolism; specifically, we inhibited cell-substrate interactions by plating the cells on Poly-2-hydroxyethyl methacrylate (Poly 2-HEMA)-coated dishes. This suppressed FAK phosphorylation at Y397, as expected, with concomitant reduction in glutamine utilisation in the TCA cycle (Figure S3A, B and C).
My background/expertise is in developmental and adult neurogenesis, in vivo modelling of gliomagenesis and cell fate control/reprogramming, with a focus on molecular mechanisms of differentiation and quantitative aspects of lineage dynamics; molecular details of the control of cellular metabolism, cell-cell adhesion and cytoskeletal dynamics are not core expertise of mine.
We appreciate this reviewer's expertise are not necessarily in the cancer cell biology and genetic intervention aspects of our study. We hope that the explanations we have provided satisfy the reviewer that our conclusions are valid.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Masalmeh and colleagues employ a neural stem/progenitor cell-based glioma model (NPE cells) to investigate the role of Focal Adhesion Kinase (FAK) in GBM, with a focus on potential links between the regulation of morphological/adhesive and metabolic GBM cell properties. For this, the authors employ wt cells alongside newly generated FAK-KO and -reexpressing cells, as well as pharmacological interventions to probe the relevance of specific signaling pathways. The authors´ main claims are that FAK crucially modulates glioma cell morphology, cell-cell and cell-substrate interactions and motility, as well as their metabolism, and that these effects translate to changes to relevant in vivo properties such as invasion and tumor growth. My main issues are with the model chosen by the authors.
As per the methods section, generation of FAK-KO and -"Rx" NPE cells entailed protracted selection/expansion processes, which may have resulted in inadvertent selection for cellular/molecular properties unrelated to the desired one (loss or gain of FAK expression) and which may have had cascading effects on NPE cells. The authors nonetheless repeatedly claim the parameters they quantify, such as mitochondrial or cytoskeletal properties or metabolic features, to have directly resulted from FAK loss or reintroduction. Examples of such causal inferences are to be found in lines 123, 134/135, 165, 181. Such causal claims are, in my view, unsupported. Acute perturbation of FAK expression/activity, genetically or pharmacologically, followed by a rapid assessment of the processes under investigation, would be needed to begin to assess causality, even if acute genetic perturbations may be technically challenging as sufficient gene expression reduction or restoration to physiologically relevant levels may be hard to achieve.
The employed pharmacological modulation of ROCK activity is the only approach that, given the presumably acute nature of the treatment, may have allowed the authors to probe the proposed functional links. The methods section of the manuscript does not however comprise details as to the duration of these treatments, which leaves open the possibility of long-term treatment having been carried out (data shown in Figure 5B refers to 72hr treatment). Even in the case of ROCK inhibitor experiments, it is however unclear if and how the effects on cell morphology and adhesion, mitochondrial organization and metabolic activity may be connected to each other and, if at all, to FAK expression.
Given the above uncertainties due to the nature of the model and experimental approaches, it is hard to assess the reliability and thus the relevance of the findings.
Experimental support for the ability of cell-substrate interaction modulation to concomitantly impact cellular metabolism and motility/invasion would be significant both in terms of advancing our understanding of glioma cell biology and of its translational potential, but the evidence being provided is at best compatible with the proposed model.
My background/expertise is in developmental and adult neurogenesis, in vivo modelling of gliomagenesis and cell fate control/reprogramming, with a focus on molecular mechanisms of differentiation and quantitative aspects of lineage dynamics; molecular details of the control of cellular metabolism, cell-cell adhesion and cytoskeletal dynamics are not core expertise of mine.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Review of Masalmeh et al.
Title: "FAK modulates glioblastoma stem cell energetics..."
Previous studies have implicated FAK and the related tyrosine kinase PYK2 in glioblastoma growth, cell migration, and invasion. Herein, using a murine stem cell model of glioblastoma, the authors used CRISPR to inactivate FAK, FAK-null cells selected and cloned, and lentiviral re-expression of murine FAK in the FAK-null cells (termed FAK Rx) was accomplished. FAK-/- cells were shown to possess epithelial characteristics whereas FAK Rx cells expressed mesenchymal markers and increased cell migration/invasion in vitro. Comparisons between FAK-/- and FAK Rx cells showed that FAK re-expressed increased mitochondrial respiration and amino acid uptake. This was associated with FAK Rx cells exhibiting filamentous mitochondrial morphology (potentially an OXPHOS phenotype) and decreased levels of MTFR1L S235 phosphorylation (implicated in mito morphology fragmentation). Mito and epithelial cell morphology of FAK-/- cells was reversed by treatment with Rho-kinase inhibitors that also increased mito metabolism and cell viability. Last, FAK-dependent glioblastoma tumor growth was shown by comparisons of FAK-/- and FAK Rx implantation studies.
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
Some questions that would enhance potential impact.
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
This manuscript addresses the question of whether inhibitors of the phosphatases Eya1-4 and of the kinase PLK1 provide an effective therapeutic approach to a range of cancers. Both Eyas and PLK1 have well documented roles in development, and have been implicated in a subset of tumors. Moreover, the authors have previously shown that PLK1 is a substrate of Eya phosphatase activity. Building on these previous findings, the authors assess the possibility of combining an Eya inhibitor, benzarone, with a PLK1 inhibitor, BI2536.
There are several concerns with the study: 1. The authors suggest that these two drugs are synergistic. Synergy is usually taken as indicative of a greater than additive effect of the two drugs. The ZIP synergy score tested here indicates that the combination of the two drugs has a synergy score between 0 and 10 (figure1, and figure 5). According to "Synergy Finder", "A ZIP synergy score of greater than 10 often indicates a strong synergistic effect, while a score less than -10 suggests a strong antagonistic effect. Scores between -10 and 10 are typically considered additive or near-additive." The data in figure 2 on mitotic cell fraction and on cell death also seems to be more of an additive effect of the two drugs than synergy. The data in figure 3 are also additive effects on RAD51. Therefore a conclusion that "These data indicate that the drug combination was broadly synergistic" seems unwarranted.
There is a general lack of nomenclature standardisation for defining synergy. Furthermore, multiple synergy models exist, with discrepancies between them. However, as the reviewer states, the prevailing view is that synergy is a combination effect that is stronger than the additive effect of the two drugs. Synergy scores derived from dose-response matrices using different synergy scoring models with scores that fall above 5 are considered truly synergistic (Malyutina A et al., 2019). To strengthen our conclusion of synergy between PLK1 and EYA inhibitors, we have calculated synergy scores using additional synergy models for both benzarone + BI2536 and benzarone + volasertib in H4 and T98G cell lines. Specifically, we find robust synergy (>5) using ZIP, HSA and Bliss calculations with the Benzarone + BI2536 drug combination in H4 cells and with Benzarone + Volasertib in H4 and T98G cells. Synergy scores for Benzarone + BI2536 fell just below 5 in T98G cells. These data are now included in Supplemental Fig S1G of the revised manuscript.
The discovery of synergistic drug combinations can be further strengthened by evaluating synergy across multiple cellular models. In this study, we have tested a total of 27 different cancer models that universally support synergy.
Regarding the phenotypic outcomes (mitotic cell fraction, cell death, RAD51 foci), we agree that the observed effects are additive. This is consistent with overall synergistic effects on viability being caused by a combination of additive mechanistic effects. We have amended the text in the revised manuscript to clarify this point.
There was no statistical difference in the synergy scores of the "high expressing" versus "low expressing cells". So the conclusion that the drug combination "t was effective at lower doses in cell lines with high levels of EYA1 and/or EYA4" seems unwarranted based on the data. Moreover, since there was no statistical difference in synergy between high and low expressing cells, stating that "the potential utility of the combination treatment depends on the specific overexpression of EYA1 and/or EYA4 in cancer cells," seems unwarranted by the data.
Synergy scores quantify the interaction between drugs, but do not capture absolute treatment effectiveness or dose sensitivity, both of which are crucial for therapeutic considerations. We have included the following sentence in the revised manuscript to clarify this distinction: “While synergy scores did not significantly differ between high and low EYA expressors, high EYA1/4 expression was associated with increased sensitivity to the combination treatment at lower doses, as evidenced by decreased cell viability.” We have also amended the conclusions in the Abstract and Discussion to reflect that the potential utility of the combination therapy in EYA1/4-high cancers is supported by potency rather than synergy scores alone.
Benzarone and benzbromarone and their derivatives have been shown to bind and inhibit Eya phosphatases, albeit at fairly high doses. However, these two compounds also have a number of other, unrelated targets. The only demonstration that Eyas are a target of benzarone in this study are the CETSA data in supplemental figure 1. The data here seem to represent an n of 1, with no error bars shown. Even more importantly, there is no control. Looking at the blot of actin, it seems as if there may be a benzarone- temperature effect on this protein as well. It would be very helpful to show some evidence that knockdown of Eya similarly synergizes with the PLK1 inhibitor, show data that benzarone is in fact inhibiting Eya activity in these cells by looking at known targets (ie the carboxyterminal tyrosine of H2AX), and other evidence of specificity.
The specificity of benzarone to the EYA proteins has been demonstrated previously using both in vitro phosphatase assays and the assessment of EYA-mediated pathways (Tadjuidje et al., 2012; Wang et al., 2021; Nelson et al., 2024). These publications have been cited in the manuscript. In addition, benzarone produces phenotypes consistent with the known functions of the EYAs (ie, reduction of PLK1 activity, reduction in RAD51 foci, G2/M arrest, and apoptosis). To further validate EYA target specificity, we have performed viability assays on control and EYA4-depleted HeLa, H4 and T98G cells in response to BI2536 treatment, demonstrating EYA4 depletion-mediated sensitization to BI2536. These data are now included in Fig 1H of the revised manuscript.
To strengthen our CETSA data, we have now included: (i) densitometry of actin, demonstrating a lack of benzarone-temperature effect, (ii) CETSA analysis for an additional cell line (T98G), demonstrating enhanced thermal stability of the EYAs in the presence of benzarone, and (iii) CETSA analysis of an additional protein (BUB1) to demonstrate target specificity. These data are now included in Supplemental Fig S1E and F of the revised manuscript.
The proteomic and transcriptomic data of cell lines that were vulnerable to the combination of BI2536 and benzarone implicate overall changes in chromatin with sensitivity. These findings call into question the idea that these two compounds are acting selectively on PLK1 and Eyas. The authors don't really provide any model for explaining this correlation of Nurd complex components with targeting Eyas and PLK1.
The proteomic and transcriptomic data demonstrate that sensitivity to the combination treatment is associated with higher expression of NuRD complex members and other chromatin regulators. This suggests that cell lines with certain chromatin configurations might be more susceptible to the combined inhibition of PLK1 and EYA. This does not undermine the demonstrated on-target effects of the two compounds, but rather suggests a potential contextual dependence of drug efficacy on chromatin state. Our data thereby implicate NuRD complex expression as a predictive biomarker for tumours that are likely to respond to EYA and PLK1 combination therapy. This has now been clarified in the discussion section of the revised manuscript.
Specificity of antibodies: I would like to see validation of the Eya antibodies, given the difficulty with such reagents in the field.
All EYA antibodies have now been validated by western blot analysis following siRNA-mediated depletion. These data are presented in Supplemental Fig S1A of the revised manuscript.
Reviewer #1 (Significance (Required)):
New therapies targeting glioblastoma would be welcome. It is not clear that the combination tested here is an effective approach to therapy. It would be necessary to know the targets of the combination and understand the mechanism so that the approach could be pursued further,
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
This study explores the sensitivity of cancer cell lines, particularly GBM cells, to dual inhibition of EYA and PLK1, aiming to uncover the connection between these pathways and the cancer stem cell state. Additionally, it investigates whether the NuRD complex modulates GBM cell responses to EYA and PLK1 inhibition. While the findings are interesting, further clarification is needed to establish the mechanistic links between EYA, PLK1, and NuRD, as well as a stronger rationale for their targeted inhibition in GBM therapy- this can be better clarified.
Some key comments and recommendations: The findings demonstrate that the combination of Benzarone (EYAi) and volasertib (PLKi) significantly reduced cell proliferation in H4 and T98G GBM cell lines, both of which show high expression of EYA. In contrast, the low EYA-expressing A172 cells exhibited limited response. A possible explanation is the inherently slower proliferation rate of A172 cells, which may reduce their dependence on G2/M arrest, thereby diminishing the impact of PLK1i. Does A172 line show a similar growth or cell division rate to H4 and T98G lines.
A172 cells have a slower proliferation rate than H4 or T98G cells, which may diminish their response to EYA/PLK1 inhibitors. However, in this study we have tested a total of 15 cancer cell lines and 12 GBM stem cell line models. No clear correlation between cell growth rate and sensitivity was observed. As a specific example, the low EYA expressing SJSA-1 cell line has a high proliferation rate but is a low responder to EYA1/PLK1 inhibitors.
Additionally, although protein expression levels of EYA were assessed across these cell lines, the activity and expression levels of PLK1 were not fully characterized. Since PLK1 is a crucial regulator of mitotic entry and DNA damage repair, its activity across cell lines may contribute to the observed variations in drug sensitivity. Could the authors investigate levels of PLK in these cell lines?
To address this point, we compared PLK1 expression levels across the panel of cancer cell lines used in our study. These data are now included in Supplemental Fig S1D of the revised manuscript, and show that PLK1 levels are comparable across the cell lines, indicating that baseline PLK1 abundance does not fully explain the observed differential sensitivity.
The study describes the combination treatment as synergistic in H4 and T98G cells, however this synergy is unclear in Fig 2A and Supplemental Fig S2A. The data suggest that H4 and T98G cells exhibit sensitivity to either EYA or PLK1 inhibition alone, with combined treatment showing enhanced effects rather than synergy. This distinction is evident as BI2536 alone induces robust G2/M arrest with decreased G1 and S phase cells. To validate these findings, combination treatment should be tested in additional GBM cell lines. Additionally, repeating FUCCI cell cycle assays in A172 and H4 cells, particularly in H4, where increased γH2AX and phospho-H3 were detected in response to individual inhibitors, would provide more definitive insights into treatment-induced cell cycle dynamics.
We agree that several of the phenotypic outcomes, for example G2/M arrest (Fig 2A) and micronuclei formation (Supplemental Fig S2A), produce additive rather than synergistic effects in the combination treated cells. The major claim of the study is that the combination treatment results in potent loss of cell viability in EYA1/EYA4 overexpressing cancer cell models. This is consistent with a combination of additive mechanistic effects causing overall synergistic effects on cancer cell viability. We have clarified this point in the revised manuscript.
We have previously struggled to get adequate FUCCI sensor expression in H4 cells. However, to address this point, we have quantified cell cycle phase distribution in H4 cells treated with benzarone, BI2536, and the drug combination, using our quantitative image-based cytometry data (Fig 3A, B). These data demonstrate an accumulation of H4 cells in G2/M following combination treatment, consistent with the FUCCI data from T98G cells. Cell cycle dynamics of H4 cells are now included in Supplemental Fig S2A of the revised manuscript.
A notable inconsistency: Figure 1 utilizes volasertib, whereas Figure 2 employs BI2536. Given that both inhibitors target PLK1 why these specific inhibitors were chosen for each experiment.
This is not the case. To clarify, BI2536 is used in both Fig 1 and 2. Volasertib is used in Supplemental Fig S1 to reproduce the synergy matrix, thereby demonstrating consistent results with a second PLK1 inhibitor.
The observation of increased Rad52 foci and sister chromatid exchange (SCE) upon EYA and PLK1 inhibition (Figure 3) is interesting. These findings suggest that dual inhibition impairs homologous recombination (HR), reinforcing the role of EYA and PLK1 in maintaining genomic stability.
We agree.
Figure 4 suggests that SJH1 cells, with low EYA expression, exhibit increased sensitivity to EYA inhibition - does this cell line show high expression of PLK or NuRD?
To clarify, Fig 4 shows that SJH1 cells, which display moderate levels of EYA expression, are highly sensitive to EYA/PLK1 inhibition. Consistent with the observed positive correlation between NuRD protein expression and EYA/PLK1 inhibitor sensitivity, SJH1 cells exhibit the highest levels of NuRD components relative to the other GBM stem cell lines. Expression levels of NuRD components across the slightly sensitive, moderately sensitive, and highly sensitive GBM stem cell lines from publicly available proteomic data and western blot analysis have now been included in Supplemental Fig S5A and B of the revised manuscript, further demonstrating this positive correlation.
It seems like EYA1 (HW1) and EY4 (SB2B and PB1) expression levels are better predictors of sensitivity to treatment, but not EYA2 and 3 (which is high in H4)- can the authors comment on this?
Overall, EYA1 and EYA4 expression levels are the major predictors of EYA/PLK1 inhibitor sensitivity in both the cancer cell lines (Fig 1) and the GBM stem cell models (Fig 4). EYA3 levels are also positively associated with sensitivity in the GBM stem cell models, but not in the cancer cell lines. Despite being consistently high, EYA2 expression levels were not associated with sensitivity in either model. These intricacies are likely to reflect functional differences between the proteins, and their ability to form different sub-complexes with each other. We have now clarified these points in the discussion of the revised manuscript.
Reviewer #2 (Significance (Required)):
It remains unclear whether NuRD complex involvement is independent of EYA expression levels. Since EYA and PLK1 regulate cell cycle progression and DNA repair, further investigation is needed to delineate their connection to NuRD-mediated chromatin remodeling and differentiation programs. Overall, this study provides some interesting evidence for targeting transcriptional and mitotic vulnerabilities in GBM but requires further validation of synergistic mechanisms, differential inhibitor effects, and NuRD complex involvement in regulating the EYA-PLK1 axis.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
This manuscript extends the findings of the interactions between EYA family members and PLK1. The idea to combine EYA inhibitors and PLK1 inhibitors is a thoughtful approach. The effects on proliferation and DNA damage are useful. This effort is a combination of preclinical efforts and some mechanistic efforts and will require additional efforts to support the conclusions drawn.
Major concerns: 1. The preclinical studies will absolutely require in vivo studies. All brain tumor treatments are limited by delivery across the blood-brain barrier. It is critical to have intracranial survival studies to support the significance of the findings.
In this study, we have focused on in vitro models including cancer cell lines, GBM stem cell models and 3D tumor spheroids, to establish proof-of-principle as well as mechanistic insight for combined EYA/PLK1 inhibition. We recognize that blood-brain-barrier penetration and therapeutic efficacy in vivo are key translational steps; however, we feel that benzarone is a suboptimal drug candidate for in vivo evaluation. Future development of second-generation EYA inhibitors with higher potency, improved selectivity, and better blood-brain-barrier permeability, is currently underway by ourselves and other groups. These compounds are likely to be more suitable for future in vivo studies, including pharmacokinetic profiling, blood-brain-barrier penetration assays, and orthotopic intracranial tumour models to assess their therapeutic potential more rigorously.
Likewise, cancer stem cell studies require in vivo studies.
As outlined above, we feel that in vivo studies fall beyond the scope of this study.
The proper studies of sphere formation would include in vitro limiting dilution assays. I would suggest greater depth in stem cell and differentiation marker studies to understand what the connection to stemness is.
The limiting dilution assay is used to measure the self-renewal potential of cancer stem cells, and would be used in this context to determine whether the treatments impact cellular differentiation. This is not the focus of this study. Rather, we are interested in comparing drug sensitivity in cancer stem cells versus differentiated cancer cells. Nevertheless, this is a great suggestion for future investigation as part of a more detailed evaluation of stemness and how these drugs and drug combinations impact self-renewal.
DNA damage responses differ between cancer stem cells and differentiated tumor cells. I would suggest comparison of effects between matched cells with different cell states.
We agree that cancer stem cells and their differentiated counterparts often display distinct DNA damage responses. We have tried to mimimise the impact of these differences on the overall conclusions by using multiple cancer cell lines and GBM stem models. To address this comment, we performed western blot analysis of DNA damage response proteins in matched PB1 stem cells and differentiated cells, demonstrating comparable expression of DNA damage response proteins. These data have now been included in Supplemental Fig S5C of the revised manuscript.
While the inhibitors used may have general specificity for the molecular targets, I would suggest that the authors use genetic loss-of-function and gain-of-function studies to validate the findings. It is particularly important because the primary targets do not predict treatment responses. I would suggest that rescues with PLK1 phosphorylation mutants would be helpful.
Our data demonstrate that EYA expression levels are predictive of treatment response in both cancer cell lines and GBM stem cell models. To further validate EYA target specificity, we have used a genetic loss-of-function approach. Specifically, we performed viability assays on control and EYA4-depleted HeLa, H4 and T98G cells in response to BI2536 treatment, demonstrating EYA4 depletion-mediated sensitization to BI2536. These data are now included in Fig 1H of the revised manuscript.
We have previously performed comprehensive rescue experiments with PLK1 phosphorylation mutants (Fig 5C–K; Nelson et al., Nat. Commun. 2024). These experiments demonstrated that cell death in response to EYA depletion or inhibition is attributable to the phosphorylation status of pY445 on PLK1, with an accumulation of Y445 phosphorylation reducing PLK1 activity and functionality, culminating in the potent induction of mitotic cell death.
Figure 5 should be performed with several lines across different response groups.
Our study currently includes cell viability and proliferation data from multiple models including 15 cancer cell lines and 12 GBM stem cell line models, spanning different EYA expression levels, and displaying varying sensitivities to both single agents and the EYA/PLK1 combination treatment. We then narrowed the number of models significantly for follow-up analysis. In Fig 5, we selected the highly sensitive PB1 GBM stem cell line based on its ability to form and grow as spheroids. While we appreciate the suggestion to expand these analyses to additional lines, we would like to respectfully decline growing additional spheroids at this time due to limitations inherent in the expansion of these models. We believe that the current dataset adequately demonstrates the reproducibility and relevance of our findings across different response groups.
The molecular associations are currently just associations. I would suggest greater analysis using genetic manipulation to test causation.
To address this concern, we have performed additional experiments using siRNA-mediated knockdown of EYA4 in HeLa, H4 and T98G cells. These experiments demonstrate that depletion of EYA4 sensitizes cells to PLK1 inhibition, mimicking the effects observed with pharmacological EYA inhibition. These data have been included in Fig 1H of the revised manuscript, and provide additional functional evidence supporting a causal relationship between EYA activity and sensitivity to PLK1 inhibition.
Figure 6 should be better developed to include protein testing and validation.
To address this point, expression levels of NuRD components have been compared using publicly available proteomic datasets and western blot analysis across the slightly sensitive, moderately sensitive and highly sensitive GBM stem cell lines, supporting a positive correlation with sensitivity. These data have been included in Supplemental Fig S5A and B of the revised manuscript.
Reviewer #3 (Significance (Required)):
This is a modest advance in understanding how EYA family members may function with PLK1.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript extends the findings of the interactions between EYA family members and PLK1. The idea to combine EYA inhibitors and PLK1 inhibitors is a thoughtful approach. The effects on proliferation and DNA damage are useful. This effort is a combination of preclinical efforts and some mechanistic efforts and will require additional efforts to support the conclusions drawn.
Major concerns:
The preclinical studies will absolutely require in vivo studies. All brain tumor treatments are limited by delivery across the blood-brain barrier. It is critical to have intracranial survival studies to support the significance of the findings.
Likewise, cancer stem cell studies require in vivo studies.
The proper studies of sphere formation would include in vitro limiting dilution assays. I would suggest greater depth in stem cell and differentiation marker studies to understand what the connection to stemness is.
DNA damage responses differ between cancer stem cells and differentiated tumor cells. I would suggest comparison of effects between matched cells with different cell states.
While the inhibitors used may have general specificity for the molecular targets, I would suggest that the authors use genetic loss-of-function and gain-of-function studies to validate the findings. It is particularly important because the primary targets do not predict treatment responses. I would suggest that rescues with PLK1 phosphorylation mutants would be helpful.
Figure 5 should be performed with several lines across different response groups.
The molecular associations are currently just associations. I would suggest greater analysis using genetic manipulation to test causation.
Figure 6 should be better developed to include protein testing and validation.
This is a modest advance in understanding how EYA family members may function with PLK1.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This study explores the sensitivity of cancer cell lines, particularly GBM cells, to dual inhibition of EYA and PLK1, aiming to uncover the connection between these pathways and the cancer stem cell state. Additionally, it investigates whether the NuRD complex modulates GBM cell responses to EYA and PLK1 inhibition. While the findings are interesting, further clarification is needed to establish the mechanistic links between EYA, PLK1, and NuRD, as well as a stronger rationale for their targeted inhibition in GBM therapy- this can be better clarified.
Some key comments and recommendations:
The findings demonstrate that the combination of Benzarone (EYAi) and volasertib (PLKi) significantly reduced cell proliferation in H4 and T98G GBM cell lines, both of which show high expression of EYA. In contrast, the low EYA-expressing A172 cells exhibited limited response. A possible explanation is the inherently slower proliferation rate of A172 cells, which may reduce their dependence on G2/M arrest, thereby diminishing the impact of PLK1i. Does A172 line show a similar growth or cell division rate to H4 and T98G lines.
Additionally, although protein expression levels of EYA were assessed across these cell lines, the activity and expression levels of PLK1 were not fully characterized. Since PLK1 is a crucial regulator of mitotic entry and DNA damage repair, its activity across cell lines may contribute to the observed variations in drug sensitivity. Could the authors investigate levels of PLK in these cell lines?
The study describes the combination treatment as synergistic in H4 and T98G cells, however this synergy is unclear in Figure 2A and EV 2A. The data suggest that H4 and T98G cells exhibit sensitivity to either EYA or PLK1 inhibition alone, with combined treatment showing enhanced effects rather than synergy. This distinction is evident as BI2536 alone induces robust G2/M arrest with decreased G1 and S phase cells. To validate these findings, combination treatment should be tested in additional GBM cell lines. Additionally, repeating FUCCI cell cycle assays in A172 and H4 cells, particularly in H4, where increased γH2AX and phospho-H3 were detected in response to individual inhibitors, would provide more definitive insights into treatment-induced cell cycle dynamics.
A notable inconsistency: Figure 1 utilizes volasertib, whereas Figure 2 employs BI2536. Given that both inhibitors target PLK1 why these specific inhibitors were chosen for each experiment.
The observation of increased Rad52 foci and sister chromatid exchange (SCE) upon EYA and PLK1 inhibition (Figure 3) is interesting. These findings suggest that dual inhibition impairs homologous recombination (HR), reinforcing the role of EYA and PLK1 in maintaining genomic stability.
Figure 4 suggests that SJH1 cells, with low EYA expression, exhibit increased sensitivity to EYA inhibition - does this cell line show high expression of PLK or NuRD?
It seems like EYA1 (HW1) and EY4 (SB2B and PB1) expression levels are better predictors of sensitivity to treatment, but not EYA2 and 3 (which is high in H4)- can the authors comment on this?
It remains unclear whether NuRD complex involvement is independent of EYA expression levels. Since EYA and PLK1 regulate cell cycle progression and DNA repair, further investigation is needed to delineate their connection to NuRD-mediated chromatin remodeling and differentiation programs.
Overall, this study provides some interesting evidence for targeting transcriptional and mitotic vulnerabilities in GBM but requires further validation of synergistic mechanisms, differential inhibitor effects, and NuRD complex involvement in regulating the EYA-PLK1 axis.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript addresses the question of whether inhibitors of the phosphatases Eya1-4 and of the kinase PLK1 provide an effective therapeutic approach to a range of cancers. Both Eyas and PLK1 have well documented roles in development, and have been implicated in a subset of tumors. Moreover, the authors have previously shown that PLK1 is a substrate of Eya phosphatase activity. Building on these previous findings, the authors assess the possibility of combining an Eya inhibitor,benzarone, with a PLK1 inhibitor, BI2536.
There are several concerns with the study:
The authors suggest that these two drugs are synergistic. Synergy is usually taken as indicative of a greater than additive effect of the two drugs. The ZIP synergy score tested here indicates that the combination of the two drugs has a synergy score between 0 and 10 (figure1, and figure 5) . According to "Synergy Finder" , "A ZIP synergy score of greater than 10 often indicates a strong synergistic effect, while a score less than -10 suggests a strong antagonistic effect. Scores between -10 and 10 are typically considered additive or near-additive." The data in figure 2 on mitotic cell fraction and on cell death also seems to be more of an additive effect of the two drugs than synergy. The data in figure 3 are also additive effects on RAD51. Therefore a conclusion that "These data indicate that the drug combination was broadly synergistic" seems unwarranted. Indeed, the data form
There was no statistical difference in the synergy scores of the "high expressing" versus "low expressing cells". So the conclusion that the drug combination "t was effective at lower doses in cell lines with high levels of EYA1 and/or EYA4" seems unwarranted based on the data. Moreover, since there was no statistical difference in synergy between high and low expressing cells, stating that "the potential utility of the combination treatment depends on the specific overexpression of EYA1 and/or EYA4 in cancer cells," seems unwarranted by the data.
Benzarone and benzbromarone and their derivatives have been shown to bind and inhibit Eya phosphatases, albeit at fairly high doses. However, these two compounds also have a number of other, unrelated targets. The only demonstration that Eyas are a target of benzarone in this study are the CETSA data in supplemental figure 1. The data here seem to represent an n of 1, with no error bars shown. Even more importantly, there is no control. Looking at the blot of actin, it seems as if there may be a benzarone- temperature effect on this protein as well. It would be very helpful to show some evidence that knockdown of Eya similarly synergizes with the PLK1 inhibitor, show data that benzarone is in fact inhibiting Eya activity in these cells by looking at known targets (ie the carboxyterminal tyrosine of H2AX), and other evidence of specificity.
The proteomic and transcriptomic data of cell lines that were vulnerable to the combination of BI2536 and benzarone implicate overall changes in chromatin with sensitivity. These findings call into question the idea that these two compounds are acting selectively on PLK1 and Eyas. The authors don't really provide any model for explaining this correlation of Nurd complex components with targeting Eyas and PLK1.
Specificity of antibodies: I would like to see validation of the Eya antibodies, given the difficulty with such reagents in the field.
New therapies targeting glioblastoma would be welcome. It is not clear that the combination tested here is an effective approach to therapy. It would be necessary to know the targets of the combination and understand the mechanism so that the approach could be pursued further,
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
We would like to thank the reviewers for taking the time to review our manuscript and for providing valuable comments on how to improve it. We are pleased to see that both reviewers recognize the novelty and importance of our study, its conceptual advance and potential clinical significance. They also noted the novelty and value of our functional mechanistic approach using epigenetic editing. Below, we provide a point-by-point response to their questions and points raised. The changes introduced in response to their feedback are highlighted in yellow in the revised manuscript file.
__Reviewer #1 (Evidence, reproducibility and clarity (Required)): __
Summary This study by Prada et al. aimed to explore DNA methylation and gene expression in primary EpCAMhigh/PDPNlow cells, consisting of for (probably) the largest part of AT2 cells, to understand the molecular mechanisms behind the impaired regeneration of alveolar epithelial progenitor cells in COPD. They found that higher or lower promoter methylation in COPD-associated cells was inversely correlated with changes in gene expression, with interferon signaling emerging as one of the most upregulated pathways in COPD. IRF9 was identified as the master regulator of interferon signaling in COPD. Targeted DNA demethylation of IRF9 in an A549 cell line resulted in a robust activation of its downstream target genes, including OAS1, OAS3, PSMB8, PSMB9, MX2 and IRF7, demonstrating that demethylation of IRF9 is sufficient to activate the IFN signaling pathway, validating IRF9 as a master regulator of IFN signaling in (alveolar) epithelial cells.
Major comments:
To remove airways (and blood vessels) completely from the lung tissue is difficult, if not impossible. This means that the assumption that the sorted EpCAMpos/PDPNlow cells primarily consisted of AT2 cells remains valid only if a quantitative analysis is conducted on the proportion of HT2-280pos cells in all samples in cytospins to exclude any significant contamination from bronchial epithelial cells. If authors cannot demonstrate >95% pure HT-280-positive cells, then the key conclusions suggesting that the epigenetic regulation of the IFN pathway might be crucial in AT2 progenitor cell regeneration could also potentially apply to bronchial progenitor cells. In addition, if >95% purity cannot be demonstrated, the data should be adjusted to account for differences in cell type composition.
__Response: __
We thank the reviewer for raising this important point. Although, as pointed out by the reviewer, we cannot guarantee that our sorted cells do not contain a minor contamination from respiratory / terminal bronchial cells, we carefully selected donors, tissue regions, and sorting strategy to ensure the highest possible enrichment of AT2 cells, as we explain below. We have now expanded the methods and results section and covered this point in the manuscript discussion.
AT2 marker genes (ABCA3, LPCAT1, LAMP3 and the surfactant genes SFTPA2, SFTPB and SFTPC) were among the top highly expressed genes in our RNA-seq data and were not significantly changed in COPD (please see expression data in __ S2A__ in the manuscript, and below for convenience), as well as Table 6, providing further evidence that the sorted cells carry a strong AT2 transcriptional signature. Fig. 1G* FACS plot examples showing the analysis of sorted AT2 cells (back sorting) from control (blue) and COPD (green) donors displayed over total cell lung suspensions (grey) H Representative IF staining of HT2-280 expression in sorted AT2 cells from no COPD (top) and COPD (bottom) donors. Nuclei (blue) were stained with DAPI, scale bars=20µm __Fig. S2A __Normalized read counts from RNA-seq data for AT2-specific genes in sorted AT2 cells from each donor (dots). Data points represent normalised counts from no COPD (blue), COPD I (light green) and COPD II-IV (dark green). Group median is shown as a black bar. *
In agreement with a previous study which profiled bulk AT2 using expression arrays (PMID: 23117565), we also observed upregulation of IFN signaling pathway in COPD AT2s. The enrichment of IFNα/β signature was also observed in COPD in the inflammatory AT2 cluster (iAT2) in a recent scRNA-seq study (PMID: 36108172). As part of the revision, we compared the IFN gene signature identified in our bulk AT2 RNA-seq with a recent scRNA-seq study (published after the submission of our manuscript, PMID: 39147413) that profiled EpCAMpos cells from COPD and non-smoker donor lungs. We observed an upregulation of our IFN signature genes in AT2 in COPD (mostly in AT2c and rbAT2 subsets), suggesting that similar signatures were observed in COPD AT2s in this dataset as well (please see __ S4E-F__ below). ____Figure S4E Expression values for the indicated genes of the IFN pathway from an external scRNA-seq dataset of AT2 cells from COPD patients and healthy controls (Hu et al, 2024). Y-axis shows log-normalized gene expression levels. F. Combined gene set score of the genes shown in (E) in different subsets of AT2 cells from Hu et al, 2024. The IFN signature genes were identified in our integrative analysis of TWGBS and RNA-seq in sorted AT2 cells.
We have also carefully examined DNA methylation profiles across all samples. The density plots of our T-WGBS DNA methylation data are very similar among the individual samples in all 3 groups, indicating that the sorted cells consist mostly of a single cell type, as there are no obvious intermediate (25-75%) methylation peaks, as observed in cell mixtures ( 2A and the panel below). No reference DNA methylation profiles are available for respiratory or terminal bronchial cells; hence, we cannot compare how epigenetically different these cells would be from AT2 nor perform a deconvolution for potential minor contamination with distal airway cells. *Figure: DNA methylation density plots of sorted EpCAMpos/PDPNneg cells from no COPD (blue, n=3), COPD I (light green, n=3) and COPD II-IV (dark green, n=5) showing a homogeneous methylation pattern and low abundance at intermediate (25%-75%) methylation values across all profiled samples, indicating that the sorted cells were mostly of a single cell type. *
We have now added a sentence to the limitations section of the discussion to cover that point specifically. CHANGES IN THE MANUSCRIPT:
AT2 cells were isolated by fluorescence-activated cell sorting (FACS) from cryopreserved distal lung parenchyma, depleted of visible airways and vessels of three no COPD controls, three COPD I and five COPD II-IV patients as previously described (24, 52, 53)
The isolated cells were positive for HT2-280, a known AT2 marker (54)*, as confirmed by immunofluorescence (Fig. 1H), validating the identity and high enrichment of the isolated AT2 populations. ** *
*Known AT2-specific genes, including ABCA3, LAMP3 and surfactant genes (SFTPA2, SFTPB and SFTPC) were among the top highly expressed genes and were not significantly changed in COPD AT2s (Fig. S2A, Table 6), further confirming the AT2-characteristic transcriptional signature of our isolated cells. *
However, 5-AZA is a global demethylating agent, and the observed effects may not be direct. To validate the epigenetic regulation of central AT2 pathways further, we took advantage of locus-specific epigenetic editing technology *(73). We focused on the IFN pathway because it was the most significantly enriched Gene Ontology (GO) term in our integrative analysis of TWGBS and RNA-seq data. Several IFN pathway members had associated hypomethylated DMRs within promoter-proximal regions and concomitant increased gene expression (Fig. 4C and S2C). Additionally, we confirmed the elevated expression of IFN-related genes with associated DMRs identified in our study in AT2 cells and AT2 cell subclusters from a recently published scRNA-seq cohort (74) (Fig. S4E-F). *
We observed upregulation of multiple IFN genes in AT2 in COPD, consistent with a previous expression array study (24). IFNα/β signaling was also enriched in COPD patients in the inflammatory AT2 cluster (iAT2) in a recent scRNA-seq study (84) and our INF signature genes were also upregulated in AT2c and AT2rb subsets in COPD, identified by another scRNA-seq study recently (74)*. ** *
Finally, despite careful removal of airways from distal lung tissue using a dissecting microscope, we cannot exclude the presence of some terminal/respiratory bronchiole cells in our FACS-isolated EpCAMpos/PDPNlow population. Recent scRNA-seq studies provided an unprecedented resolution and identified several epithelial subpopulations and transitional cells residing in the terminal/respiratory bronchioles and alveoli, including respiratory airway secretory cells (93), terminal airway-enriched secretory cells (28), terminal bronchiole-specific alveolar type-0 (AT0) (70), and emphysema-specific AT2 cells (74). These cells may contribute to alveolar repair in healthy and COPD lungs; however, with our bulk DNA methylation and RNA-seq study, we are unable to resolve all these subpopulations. Future development of single-cell methylation and non-reference-based algorithms for DNA methylation deconvolution will enable deeper epigenetic phenotyping of specific AT2 and bronchiolar cell subsets.
(Methods) Validation of IFN gene upregulation in a published scRNA-seq dataset
scRNA-seq data from (74), generously provided by M. Köningshoff, were processed using the default Seurat workflow (117). Expression of IFN-related genes was extracted and plotted as log-normalised gene expression levels in AT2 cells from control and COPD donors. Seurat's AddModuleScore() function was used to compute a gene set score for a custom IFN program using the genes listed in __Fig. S4E __and to analyse the IFN gene set scores in AT2 cell subclusters identified in (74). Briefly, average gene expression scores were computed for the gene set of interest, and the expression of control features (randomly selected) was subtracted as described in (118).
Fig. S4E and F: E. Expression values for the indicated genes of the IFN pathway from an external scRNA-seq dataset of AT2 cells from COPD patients and healthy controls (74). Y-axis shows log-normalized gene expression levels. F. Combined gene set score of the genes shown in (E) in different subsets of AT2 cells from (74). The IFN signature genes were identified in our integrative analysis of TWGBS and RNA-seq in sorted AT2 cells.
The overrepresentation of several keratins (KRT5, KRT14, KRT16, KRT17), mucins (MUC12, MUC13, MUC16, MUC20) and the transcription factor FoxJ1 is now attributed by the authors to a possible dysregulation of AT2 identity and differentiation in COPD (lines 282 - 284) where they cite refs 28, 69, 70. Authors try to support this with IF double stains for KRT5 and HT-280 to identify co-expression of KRT5 and HT2-280 in lung tissue (Figure S2H). However, the evidence for the co-expression of both markers could be presented more convincingly.
__Response: __
We found the potential co-expression of airway and alveolar markers in COPD lungs interesting and hence included it in the original manuscript. The initial discovery came from our bulk RNA-seq data, where we observed upregulation of several genes typically found in more proximal airways in COPD (mentioned above by the reviewer). Of note, some of them (e.g., FoxJ1) are expressed at very low levels. Following reviewer's comments, to validate possible colocalization of AT2 and airway markers on protein level, we performed further IF analysis. We took Z-stack images to demonstrate the co-localization of HT2-280 and Krt5 more convincingly and co-stained the same tissue regions with SCGB3A2 (a TASC/distal airway cell marker, PMID 36796082). Even though these are rare events, we were able to reproduce the existence of HT2-280/Krt5 positive, SCGB3A2 negative cells in the alveoli of COPD patients on the protein level (__Fig. S2H __and panels below). Although interesting, we decided to keep this finding in the supplement and did not include it in the discussion to focus the story on the epigenetic regulation of the IFN pathway, which is the main discovery of our study. We will investigate this observation in future studies.
Figure S2H and here: Examples of HT2-280/Krt5 double positive cells. Top, immunofluorescence staining of the alveolar region of a COPD II donor showing the existence of AT2 cells (HT2-280 positive (red), which are SCGB3A2 negative (green, left) but KRT5 positive (green, right). In conclusion, double-positive HT2-280/KRT5 cells are rare but present in the alveoli of COPD patients. Magnification: 20x. Scale bar: 50 µm. Bottom, Z-stack images highlighting HT2-280 (red) and KRT5 (green) double-positive cells at 63x magnification. Scale bar: 5 µm.
CHANGES IN THE MANUSCRIPT:
In addition, we observed an upregulation of several keratins (KRT5, KRT14, KRT16, KRT17) and mucins (MUC12, MUC13, MUC16, MUC20), suggesting a potential dysregulation of alveolar epithelial cell differentiation programs in COPD (Table 6, Fig. S2F). Immunofluorescence staining confirmed the presence of KRT5-positive cells in the distal lung in COPD and identified cells positive for both KRT5 and HT2-280 (Fig. S2H). Collectively, these results indicate a dysregulation of stemness and identity in the alveolar epithelial cells in COPD.
Fig. S2H legend: The zoomed-in panel (right corner, bottom) demonstrates the presence of rare HT2-280/KRT5 double-positive cells in the alveoli of COPD patients.* Slides were counterstained with DAPI, scale bars = 50µm, 20µm or 5µm, as displayed in images. *
Double staining for KRT5 and HT2-280 did highlight the proximity of both cell types in lung tissue, underscoring the challenge of removing airways (including the smaller and terminal bronchi) from the tissue. In addition, HT-280/KRT5 co-expression is not consistent with recent studies from refs 28, 69, 70 where other markers for distal airway cell transition, such as SCGB3A2 and BPIFB1, have been demonstrated, which were not investigated in this study.
Response:
We provided a general overview of the different signatures observed in our data, but we could not validate every deregulated pathway or gene. We include the relevant tables detailing all differentially expressed genes and differentially methylated regions to enable and encourage the community to follow up on the data in subsequent studies.
As demonstrated above, we detect the co-occurrence of HT2-280/KRT5 staining on the protein level in the same cells in the alveoli of COPD patients. We would like to emphasize that alveolar epithelial cell identity in CODP lungs has not been investigated in detail on the protein or RNA level, and HT2-280/KRT5 co-expression/co-localization has not been directly tested in the studies mentioned by the reviewer since, among other reasons, the gene encoding HT2-280 has not been identified. Notably, a recent study (published after the submission of our manuscript) focusing on enriched epithelial cells from the distal lungs of COPD patients (PMID 35078977), identified an emphysema-specific AT2 subtype co-expressing the AT2 marker SFTPC and distal airway cell transition marker SCGB3A2, indicating that disease-specific AT2 populations with possible co-occurrence of AT2 and airway markers exist. In our dataset, SCGB3A2 was not deregulated (log2 fold change=0.22, adj p-value= 0.47), as shown in Table 6, and the HT2-280/Krt5 positive cells were negative for SCGB3A2 in our IF staining (see above).
BPIFB1 is one of the antimicrobial peptides genes with an associated DMR and is significantly upregulated in COPD cells in our study (log2 fold change=1.17, adj p-value=0.0016), as shown in the supplementary figure Fig S4C and here below for convenience.
Figure S4C Fold-change in gene expression of BPIFB1 in AT2 cells in COPD (RNA-seq) and A549 cells treated with 0.5µM AZA (RT-qPCR) compared to control samples. Left, RNA-seq data from AT2 cells (no COPD, blue, n=3; COPD II-IV, green, n=5). Right, A549 treated with AZA (orange, n=3) compared to control DMSO-treated cells (grey, n=3). The group median is shown as a black bar.
The small (and not evenly divided) sample size of both COPD and non-COPD specimens may lead to a higher risk for false positive results as adjustments for multiple testing typically rely on the number of comparisons, and small sample sizes may not provide enough data points to adequately control for this.
__Response: __
We acknowledge the problem of testing for multiple traits with relatively small numbers of samples. The availability of donor tissue, especially from non-COPD and COPD-I donors, was limited, and we applied very strict donor matching and quality control criteria for sample inclusion to avoid additional variability and confounding factors. The importance of strict quality control in selecting appropriate control samples was highlighted in our previous study (PMID: 33630765), where we demonstrated that approximately 50% of distal lung tissue from cancer patients with normal spirometry has pathological changes. Hence, we believe that the quality of the tissue was paramount to the reliability of the data. Strict quality control and sample matching for multiple parameters, including age, BMI, smoking status and smoking history (critical for DNA methylation studies), and cancer type (for background tissue), is a key strength of our approach, but it inevitably limited our sample size.
First, all samples were cryopreserved and then processed in parallel in groups of 1 non-COPD and 2-3 COPD samples. This process included tissue dissociation, FACS sorting, back sorting (always), and immunofluorescence staining (when enough material was available). Cell pellets were stored at -80{degree sign}C until the entire cohort was ready for sequencing. This was done to limit the potential variation introduced by processing and sorting. RNA and DNA isolations were performed in parallel for all the sorted cell pellets, which were then sequenced as a single batch.
During data analysis, we applied stringent cutoffs for DMR detection to reduce the risk of false positives due to multiple comparisons and a small sample size. Specifically, we filtered for regions with at least 10% methylation difference and containing at least 3 CpGs. Additionally, we applied a non-parametric Wilcoxon test using average DMR methylation levels to remove potentially false-positive regions, as the t-statistic is not well suited for non-normally distributed values, as expected at very low/high (close to 0% / 100%) methylation levels. A significance level of 0.1 has been used. Therefore, we are confident that the rigorous analysis and strict criteria applied in this study allowed us to detect trustworthy DMRs that we could further functionally validate using epigenetic editing. All the details of the DMR analysis are provided in the methods section. To address this point and limitation, we have added the following paragraphs in the discussion section of the manuscript:
CHANGE IN THE MANUSCRIPT:
*The strengths of our study include the use of purified human alveolar type 2 epithelial progenitor cells from a well-matched and carefully validated cohort of human samples, including mild and severe COPD patients, providing high relevance to human COPD. *
However, we acknowledge several limitations of our study that warrant further investigation. First, the sample size was small. The use of strict quality criteria for donor selection limited the available samples, particularly for the ex-smoker control group. This resulted in an unequal distribution of COPD and control samples. This impacts the power of statistical analysis, particularly in the WGBS analysis, where millions of regions genome-wide are tested. Nevertheless, the clear negative correlation between promoter methylation and corresponding gene expression highlights the robustness of the DMR selection. Additionally, we were able to experimentally validate interferon-associated DMRs using epigenetic editing, highlighting the power of integrated epigenetic profiling in identifying disease-relevant regulators.
__Minor suggestions for improvement __
__Introduction __ • In general, refer to the actual experimental studies rather than review papers where appropriate.
Response:
We have now carefully checked all the references and amended them to refer to experimental studies when required.
Clearly specify whether a study was conducted in mice or humans, as this distinction is crucial for understanding the relevance of the findings to COPD.
__Response: __
All our experiments were performed with human lung cells and tissues. No mouse samples were used. As suggested, we have now clearly stated that our study was performed using human tissue samples and cells in different parts of the manuscript, including the discussion, where we now explicitly highlight the strengths and limitations of our study.
CHANGES IN THE MANUSCRIPT:
...we generated whole-genome DNA methylation and transcriptome maps of sorted human primary alveolar type 2 cells (AT2) at different disease stages.
However, the regulatory circuits that drive aberrant gene expression programs in human AT2 cells in COPD are poorly understood
Therefore, we set out to profile DNA methylation of human AT2 cells at single CpG-resolution across COPD stages.
...*suggesting that aberrant epigenetic changes may drive COPD phenotypes in human AT2. *
To identify genome-wide DNA methylation changes associated with COPD in purified human AT2 cells...
The similarity of the methylation and gene expression profiles in the PCAs suggested that epigenetic and transcriptomic changes in human AT2 cells during COPD might be interrelated ...
*In this work, we demonstrate that genome-wide DNA methylation changes occurring in human AT2 cells may drive COPD pathology by dysregulating key pathways that control inflammation, viral immunity and AT2 regeneration. *
*Using high-resolution epigenetic profiling, we uncovered widespread alterations of the DNA methylation landscape in human AT2 cells in COPD that were associated with global gene expression changes. *
*Currently, it is unclear how cigarette smoking leads to changes in DNA methylation patterns in human AT2 *
The strengths of our study include the use of purified human alveolar epithelial progenitor cells from a well-matched and carefully validated cohort of human samples, including mild and severe COPD patients, providing high relevance to human COPD.
__Methods __ • Line 473, here is meant 3 ex-smoker controls instead of smoker controls?
__Response: __
All donors (no COPD and COPD) used in our study are ex-smokers. Matching the samples with regard to smoking status and history is critical for epigenetic studies, as cigarette smoke profoundly affects DNA methylation genome-wide (PMID: 38199042, PMID: 27651444). This has now been clarified in the revised manuscript.
CHANGE IN THE MANUSCRIPT____:
Of note, we included only ex-smokers in our profiling to avoid acute smoking-induced inflammation as a confounding factor (50)*. *
Importantly, we matched the smoking status and smoking history of all donors, which is key in epigenetic studies, as cigarette smoking profoundly impacts the DNA methylation landscape of tissues (96).
In total, 3 ex-smoker controls (no COPD), 3 mild COPD donors ex-smokers (GOLD I, COPD I) and 5 moderate-to-severe COPD donors ex-smokers (GOLD II-IV, COPD II-IV) were profiled (Fig. 1A-C, Table 1)
__Discussion __ • A list of limitation should be added to the discussion. One is the use of the alveolar cell line A549, which produces mucus, a characteristic more commonly associated with bronchial epithelial cells. (ref 43)l530:
__Response: __
The profiling was performed using purified primary human alveolar epithelial progenitor cells. For technical reasons, A549 cells were only used for validation of the results using epigenetic editing. The A549 phenotype depends on the growth medium used, in our case, Ham's F-12 medium, which is recommended for long-term A549 culture and promotes multilamellar body formation and differentiation toward an AT2-like phenotype (PMID: 27792742)__. __We are developing epigenetic editing technology for use in primary lung cells; however, the approach currently relies on the high efficiency of transient transfections, which cannot yet be achieved with primary adult AT2 cells. We were positively surprised by how well the methylation data obtained from patient AT2s translated into mechanistic insights when using A549 cells, despite being a cancer cell line. This suggests that the fundamental mechanisms of epigenetic regulation of IRF9 and the IFN signaling pathway are conserved between A549 and primary AT2 cells.
Another limitation to consider is that cells were isolated primarily from individuals with lung cancer, except for patients with COPD stage IV. In particular as COPD stage II and IV samples were taken together. And discuss the small and unevenly divided sample size
__Response: __
We thank the reviewer for bringing up this important point, which we carefully considered when designing our study. To match our samples across the cohort, all the no-COPD, COPD I, and two of the COPD II-IV samples were obtained from cancer resections. In addition to other characteristics, like age, BMI and smoking status, we also matched the donors by cancer type (all profiled donors had squamous cell carcinoma). We collected lung tissue as far away from the carcinoma as possible and sent representative pieces for histological analysis by an experienced lung pathologist to confirm the absence of visible tumours. In addition, to ensure that our data represents COPD-relevant signatures, we intentionally included samples from three COPD donors undergoing lung resections (without a cancer background) in the profiling.
Following the reviewer's suggestion, to investigate the potential impact of non-cancer samples on driving the observed differences, we carefully checked the PCAs for both DNA methylation and RNA-seq. We could not identify a clear separation of no-cancer COPD samples from the cancer COPD samples (or other cancer samples) in any examined PCs, indicating no cofounding effect of cancer background in the samples. We observed that one sample contributing to PC2 is a non-cancer sample, but this was a rather sample-specific effect, as the other two non-cancer samples clustered together with the other severe COPD samples with a cancer background. Notably, in our DNA methylation data, we do not observe typical features of cancer methylomes, like global loss of DNA methylation or aberrant methylation of CpG islands (e.g., in tumour suppressor genes) (see Fig 2A), further suggesting that we do not "pick up" confounding cancer signatures in our data.
Following the comments from both reviewers, to clarify that point, we added the information about cancer and non-cancer samples to the PCA figures for DNA methylation (new Fig. 2B) and RNA-seq (new Fig. 3A) data in the revised manuscript, as shown below
CHANGE IN THE MANUSCRIPT____:
COPD samples from donors with a cancer background clustered together with the COPD samples from lung resections, confirming that we detected COPD-relevant signatures (Fig. 2B).
Fig.2B* Principal component analysis (PCA) of methylation levels at CpG sites with > 4-fold coverage in all samples. COPD I and COPD II-IV samples are represented in light and dark green triangles, respectively, and no COPD samples as blue circles. COPD samples without a cancer background are displayed with a black contour. The percentage indicates the proportion of variance explained by each component. *
Unsupervised principal component analysis (PCA) on the top 500 variable genes revealed a clear influence of the COPD phenotype in separating no COPD and COPD II-IV samples, as previously observed with the DNA methylation analysis, irrespective of the cancer background of COPD samples (Fig.3A, Fig. S2B).
*Principal component analysis (PCA) of 500 most variable genes in RNA-seq analysis. PCA 1 and 2 are shown in Fig.3A, PCA 1 and 4 in Fig.S2B. COPD I and COPD II-IV samples are represented in light and dark green triangles, respectively, and no COPD samples as blue circles. COPD samples without a cancer background are displayed with a black contour. The percentage indicates the proportion of variance explained by each component. *
__Response: __
We thank the reviewer for suggestions on how to improve the discussion of our manuscript. We have now added a strength/limitation section to our discussion and included the points suggested by both reviewers.
CHANGE IN THE MANUSCRIPT____:
The strengths of our study include the use of purified human alveolar epithelial progenitor cells from a well-matched and carefully validated cohort of human samples, including mild and severe COPD patients, providing high relevance to human COPD. Importantly, we matched the smoking status and smoking history of all donors, which is key in epigenetic studies, as cigarette smoking profoundly impacts the DNA methylation landscape of tissues (96). With the first genome-wide high-resolution methylation profiles of isolated cells across COPD stages, we offer novel insights into the epigenetic regulation of gene expression in epithelial progenitor cells in COPD, expanding our understanding of how alterations in regulatory regions and specific genes could contribute to disease development. We identified IRF9 as a key IFN transcription factor regulated by DNA methylation. Notably, by targeting IRF9 through epigenetic modifications, we modulated the activity of the IFN pathway, which plays a crucial role in the immune response and lung tissue regeneration. Epigenetic editing techniques could offer a novel therapeutic strategy for COPD by downregulating IFN pathway activation and promoting the regeneration of epithelial progenitor cells in the lungs. Further preclinical and clinical studies are needed to validate the efficacy and safety of epigenetic editing approaches in COPD treatment (33)*. *
*However, we acknowledge several limitations to our study that warrant further investigation. First is the small sample size and replication difficulty due to the lack of available data, common challenges for studies working with sparse human material and hard-to-purify cell populations. The use of strict quality criteria in donor selection limited the available samples, especially for the ex-smoker control group, leading to an unequal distribution of COPD and control samples. Overall, this impacts the power of statistical analysis, especially in the WGBS analysis, where millions of regions genome-wide are tested. Nevertheless, the clear negative correlation of promoter methylation to the corresponding gene expression highlights the robustness of the DMR selection. Furthermore, we could experimentally validate interferon-associated DMRs using epigenetic editing, highlighting the power of integrated epigenetic profiling for the discovery of disease-relevant regulators. *
Overall, we detected a higher number of correlated DMR-DEG associations using our simple promoter-proximal linkage compared to the GeneHancer approach. Assigning enhancers to their target genes with high confidence is a complex and challenging task. Enhancers are often located far from the genes they regulate and can interact with their target genes through three-dimensional chromatin loops. Furthermore, enhancers can operate in a highly context-dependent manner, with the same enhancer regulating different genes depending on the cell type, developmental stage, or environmental signals. Determining which enhancer is active under specific conditions remains a hurdle in the field, especially since the AT2-specific chromatin profiles of enhancer marks are not yet available.
In addition, while WGBS provides unprecedented resolution and high coverage of the DNA methylation sites across the genome, it does not allow distinguishing 5-methylcytosine from 5-hydroxymethylcytosine. Therefore, we cannot exclude that some methylated sites we detected are 5-hydroxymethylated. However, as 5-hydroxymethylcytosine is present at very low levels in the lung tissue (97)*, its effect is likely marginal. *
Finally, despite careful removal of airways from distal lung tissue using a dissecting microscope, we cannot exclude the presence of some terminal/respiratory bronchiole cells in our FACS-isolated EpCAMpos/PDPNlow population. Recent scRNA-seq studies provided an unprecedented resolution and identified several epithelial subpopulations and transitional cells residing in the terminal/respiratory bronchioles and alveoli, including respiratory airway secretory cells (93), terminal airway-enriched secretory cells (28), terminal bronchiole-specific alveolar type-0 (AT0) (70), and emphysema-specific AT2 cells (74). These cells may contribute to alveolar repair in healthy and COPD lungs; however, with our bulk DNA methylation and RNA-seq study, we are unable to resolve all these subpopulations. Future development of single-cell methylation and non-reference-based algorithms for DNA methylation deconvolution will enable deeper epigenetic phenotyping of specific AT2 and bronchiolar cell subsets.
__References __ • Check references. For instance, there is no reference in the text to ref 43.
Align format of references
__Response: __
We thank the reviewer for spotting this inconsistency. We have carefully checked and aligned the format of all references. The (old) reference 43 is now mentioned in the discussion part.
__Reviewer #1 (Significance (Required)): __
The strength of this study lies in its focus on the molecular mechanisms underlying the impaired regeneration of epithelial progenitor cells in COPD. The discovery of IRF9, which regulates IFN signaling and is prominently upregulated in COPD, together with the convincing validation of the epigenetic control of the IFN pathway by targeted DNA demethylation of the IRF9 gene, adds significant value to the COPD research field.
Main limitations of the study are the relatively small sample size of both COPD and non-COPD specimens and the claim that the sorted EpCAMpos/PDPNlow cells primarily consisted of AT2 cells.
__- Describe the nature and significance of the advance (e.g. conceptual, technical, clinical) for the field. __
The nature and significance of the advance in epigenetic editing of IRF9 in COPD can be described as both conceptual and potentially clinical:
Conceptual Advance: The epigenetic editing of IRF9 enhances our understanding of the molecular mechanisms underlying COPD pathogenesis. By targeting IRF9 through epigenetic modifications, researchers were able to modulate the activity of the IFN pathway, which plays a crucial role in the immune response and lung tissue regeneration. This approach offers insights into the epigenetic regulation of gene expression in epithelial progenitor cells in COPD and expands our understanding of how alterations in specific gene methylation could contribute to disease progression.
Clinical Significance: The potential clinical significance of epigenetic editing of IRF9 lies in its implications for COPD therapy. If successful, epigenetic editing techniques could offer a novel therapeutic strategy for COPD by downregulating IFN pathway activation and promoting regeneration of epithelial progenitor cells in the lungs. Obviously, further preclinical and clinical studies are needed to validate the efficacy and safety of epigenetic editing approaches in COPD treatment.
__Response: __We thank the reviewer for recognising the importance of our study, its conceptual advance and potential clinical significance. We are pleased to see that the reviewer highlights the promise of epigenetic editing in both furthering our basic understanding of molecular mechanisms of chronic diseases and its future potential as a therapeutic strategy.
__- Place the work in the context of the existing literature (provide references, where appropriate). __ Few experimental papers have been published on epigenetic editing in lung diseases, with limited research available beyond the study referenced in citation 43. Song J, Cano-Rodriquez D, Winkle M, Gjaltema RA, Goubert D, Jurkowski TP, Heijink IH, Rots MG, Hylkema MN. Targeted epigenetic editing of SPDEF reduces mucus production in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2017 Mar 1;312(3):L334-L347. doi: 10.1152/ajplung.00059.2016. Epub 2016 Dec 23. PMID: 28011616.
Response:
We thank the reviewer for recognising the uniqueness and novelty of our study and the lack of research on the functional understanding of DNA methylation in the context of lung and lung diseases.
- State what audience might be interested in and influenced by the reported findings.
This study is of broad interest to researchers investigating the pathogenesis and treatment of COPD.
__- Define your field of expertise with a few keywords to help the authors contextualize your point of view. __
Expertise in: Lung pathology, Immunology, COPD, Epigenetics
- Indicate if there are any parts of the paper that you do not have sufficient expertise to evaluate. Less expertise in: Epigenetic Editing
__Reviewer #2 (Evidence, reproducibility and clarity (Required)): __
__Summary: __
This study aim to understand the molecular mechanisms underlying dysfunction in AT2 cells in COPD, by profiling bulk genome wide DNA methylation using Tagmentation-based whole-genome bisulfite sequencing (T-WGBS) and RNA sequencing in selectively sorted primary AT2 cells. The study stands out in it's sequencing breadth and use of an incredibly difficult cell population, and has the potential to add substantially to our mechanistic understanding of epigenetic contributions to COPD. A further highlight is the concluding aspect of the study where the authors undertook targeted modification of specific CpG methylation, provided direct, site-specific evidence for transcriptional regulation by CpG methylation.
Response:
We thank the reviewer for recognizing the conceptual and methodological advance of our study and for noting the value of our functional mechanistic approach.
__Major comments: __
The authors clearly show that there is DNA methylation alteration in AT2 cells from COPD individuals that links functional to gene expression at some level. However, I think the statement "to identify genome-wide changes associated with COPD development and progression..." and similar other references to disease development understanding is not accurate given the DNA methylation primary comparison is between control and moderate to severe COPD, with no temporal detail or evidence that they drive progression rather than are a result of COPD development. The paragraph starting on line 186 where this is a addressed to some extent is quite vague and doesn't really provide confidence that DNAm dysregulation occurs at an early stage in this context. This can be addressed by changing the focus/style of the text.
__Response: __
Thank you for raising this point. We agree with the reviewer that our cross-sectional study describes the association of methylation changes with either COPD I or more established disease (COPD II-IV) and that the observed changes may be either the driver or a result of COPD development. This has been clarified in the revised manuscript, and we removed the statements about disease initiation and progression. This is an important point; hence, we added an extra line to the discussion to make that clear.
__CHANGE IN THE MANUSCRIPT____: __
Therefore, we set out to profile DNA methylation of human AT2 cells at single CpG-resolution across COPD stages to identify epigenetic changes associated with disease and combine this with RNA-seq expression profiles.
To identify epigenetic changes associated with COPD, we collected lung tissue from patients with different stages of COPD,
....to identify methylation changes associated with mild disease, we included TWGBS data from AT2 isolated from COPD I patients (n=3) in the analysis.
Currently, we do not know whether the identified DNA methylation changes are the cause or the consequence of the disease process and not much is known about the correlation of DNA methylation with disease severity.
*However, our study is cross-sectional, our cohort included only 3 COPD I donors, and we did not have any follow-up data on the patients, so future large-scale profiling of mild disease (or even pre-COPD cohorts) in an extended patient cohort will be crucial for a better understanding of early disease and its progression trajectories. *
__Results comments and suggestions: __
For the integrated analysis, there is a focus on DMRs in promoters with very little analysis on other regions. The paragraph starting on line 317 describes some analysis on enhancers but is very brief, doesn't include information on how many/which DMRs were included, making it hard to interpret the impact of the 147 DMRs and 93 genes identified - is this nearly all DMRs and genes analysed or very few? A comparison to the promoter analysis would be of interest. Especially as the targeted region followed up with lovely functional assessment in the last sections is a gene body DMR, not a promoter DMR.
__Response: __
We thank the reviewer for pointing out the importance of changes in enhancers. We agree that extending the enhancer analysis is very interesting. However, assigning enhancers to their target genes with high confidence is a complex and challenging task. Enhancers are often located far from the gene they regulate, sometimes spanning hundreds of kilobases. They can interact with their target genes through three-dimensional chromatin loops, potentially bypassing nearby genes to activate more distant ones, making it difficult to confidently link specific enhancers to their target genes. Furthermore, enhancers can operate in a highly context-dependent manner. The same enhancer can regulate different genes depending on the cell type, developmental stage, or environmental signals. Another challenge is that enhancers often work in clusters or "enhancer landscapes," where multiple enhancers contribute to the regulation of a single gene. Disentangling the contribution of individual enhancers within such clusters and determining which enhancer is active under specific conditions remains an ongoing hurdle in the field, especially since the AT2-specific chromatin profiles of enhancer marks are not yet available.
One approach we tried to account for more distal regulatory regions was to assign DMRs to the nearest gene with a maximum distance of up to 100 kb using GREAT (Genomic Regions Enrichment of Annotations Tool) and simultaneously perform gene enrichment analysis of the associated genes. The old Figure S1C (now S1D) shows the top 10 enriched terms of either hyper- or hypomethylated DMRs, and Table 4 shows the full list of enriched terms. However, in this analysis, we did not integrate the results of the RNA-seq analysis. To demonstrate that we can correlate methylation with gene expression associations in this analysis, we then took a closer look at the WNT/b-catenin pathway, which contains 147 DMRs associated with 93 genes from the respective pathway (old Figure S3D, now S3G). Here, we showed that distal DMRs up to 100 kb away from the TSS show a high correlation with gene expression. We are including the two figures below for convenience:
*Left panels, functional annotation of genes located next to hypermethylated (top) and hypomethylated (bottom) DMRs using GREAT. Hits were sorted according to the binominal adjusted p-value and the top 10 hits are shown. The adjusted p-value is indicated by the color code and the number of DMR associated genes is indicated by the node size. Right panel, scatter plot showing distal DMR-DEG pairs associated with Wnt-signaling. Pairs were extracted from GREAT analysis (hypermethylated, DMR-DEG distance Following the reviewer's suggestion, we have now extended the enhancer analysis using the GeneHancer database, the most comprehensive, integrated resource of enhancer/promoter-gene associations. We used the GeneHancer version 5.14, which annotates 392,372 regulatory genomic elements (GeneHancer element) on the hg19 reference genome. Of the 25,028 DMRs, 18,289 DMRs (73% of all DMRs) coincided with at least one GeneHancer element, resulting in 19,661 DMR-GeneHancer associations. Next, we extracted the GeneHancer elements associated with protein-coding or long-non-coding RNAs genes, which left us with 2,144 DMR-GeneHancer associations. Next, we used only high-scoring gene GeneHancer associations ("Elite"), leaving 1,485 DMR-GeneHancer associations. Of those, we selected the GeneHancer elements, which are linked to genes differentially expressed in our RNA-seq analysis resulting in a final table of 376 DMR-GeneHancer associations (Table 9 DMR_DEG_GeneHancer, Tab 2). Similar to the promoter-proximal analysis, we analysed the correlation of expression and methylation changes of the DMR-GeneHancer associations, demonstrating a high number of negatively and positively correlated events (Fig.S3D). Finally, we performed the gene enrichment analysis for positively and negatively correlating genes. We detected significant GO term enrichments only for negatively correlating genes (Fig.S3E and Table 10_Enrichment_results, Tab2).
CHANGE IN THE MANUSCRIPT
To harness the full resolution of our whole-genome DNA methylation data, we extended the analysis beyond promoter-proximal regions and assessed how epigenetic changes in distal regulatory regions (enhancers) may relate to transcriptional differences in COPD. As the assignment of enhancer elements to the corresponding genes is challenging, we tried two different approaches. First, we used the GeneHancer database (72) to link DMRs to regulatory genomic elements (GeneHancer element). Of the 25,028 DMRs, 18,289 DMRs (73%) coincided with at least one GeneHancer element. Of those 2,144 DMR-GeneHancer associations were linked either to protein-coding or lncRNA genes. Next, we filtered for high-scoring gene GeneHancer associations ("Elite"), leaving 1,485 DMR-GeneHancer Elite associations. Of those, we selected the GeneHancer elements, which are linked to genes differentially expressed in our RNA-seq analysis, resulting in 376 DMR-GeneHancer associations (Table 9). Similar to the promoter-proximal analysis, we assessed the correlation of expression and methylation changes of the DMR-GeneHancer associations, demonstrating a high proportion of negatively and positively correlated events (Fig. S3E). Finally, we performed gene enrichment analysis for positively and negatively correlated genes. We detected significant GO term enrichments for negatively correlating genes only (Fig. S3F and Table 10), with the most pronounced term "regulation of tumor necrosis factor". In an alternative approach, we linked proximal and distal (within 100 kb from TSS) DMRs to the next gene using GREAT (57) (Fig S1C, Table 4) *and calculated Spearman correlation between DMRs and associated DEGs__. 147 DMRs were associated with high correlation rates with 93 genes from the WNT/β-catenin pathway (Fig. S3G)__, suggesting that DNA methylation may also drive the expression of genes of the WNT/β-catenin family. *
Figure S3E and F: E. Spearman correlation between gene expression and DMR methylation of DMRs assigned to gene regulatory elements using the GeneHancer database. F. GO-Term over-representation analysis of DEGs negatively correlated to DMRs in gene regulatory elements. The adjusted p-value is indicated by the color code and the percentage number of associated DEGs is indicated by the node size.
(Methods) For enhancer analysis, the GeneHancer database version 5.14, which annotates 392,372 regulatory genomic elements (GeneHancer element) on the hg19 reference genome, was used (72). Of the 25,028 DMRs 18,289 DMRs coincided with at least one GeneHancer element, resulting in 19,661 DMR-GeneHancer associations. Next, the GeneHancer elements were filtered for association with protein-coding or long-non-coding RNAs genes and high-scoring gene GeneHancer associations ("Elite"), leaving 1,485 DMR-GeneHancer associations. Of those, the GeneHancer elements were selected, which are linked to differentially expressed genes in COPD resulting in a final table of 376 DMR-GeneHancer associations. Similar to the promoter-proximal analysis, the Spearman correlation of expression and methylation changes of the DMR-GeneHancer associations was assessed. GO gene enrichment analysis for positively and negatively correlating genes was done using Metascape (111).
A comparison to the promoter analysis would be of interest.
Response:
We detected more highly correlated (|correlation coefficient| > 0.5) DMR-DEG associations using our simple promoter proximal linkage (n=643) in comparison with the GeneHancer approach comprising annotated enhancer elements (n=327/2,144). Gene enrichment results pointed to the interferon pathway, which we could confirm using epigenetic editing. This pathway was not present in the GeneHancer analysis, indicating that regulation of the IFN pathway may be controlled by proximal elements.
CHANGE IN THE MANUSCRIPT____:
Overall, we detected a higher number of correlated DMR-DEG associations using our simple promoter-proximal linkage compared to the GeneHancer approach. Assigning enhancers to their target genes with high confidence is a complex and challenging task. Enhancers are often located far from the genes they regulate and can interact with their target genes through three-dimensional chromatin loops. Furthermore, enhancers can operate in a highly context-dependent manner, with the same enhancer regulating different genes depending on the cell type, developmental stage, or environmental signals. Determining which enhancer is active under specific conditions remains a hurdle in the field, especially since the AT2-specific chromatin profiles of enhancer marks are not yet available.
Especially as the targeted region followed up with lovely functional assessment in the last sections is a gene body DMR, not a promoter DMR.
Response:
We thank the reviewer for bringing up that point. To clarify, we defined the promoter regions for the analysis as regions located {plus minus} 6 kb (upstream and downstream) from the transcriptional start site (TSS). Since the term "promoter" often refers to the region upstream of the transcriptional start site, its use may have been misleading. For clarity, we changed the text correspondingly to __promoter proximal methylation __and explained in the methods how the regions for analysis were defined.
__CHANGE IN THE MANUSCRIPT____: __
"DMR association per gene promoter" was changed to "Gene promoter proximal DMRs"
Fig. S3B: "DMR in promoter" was changed to "promoter proximal DMR(s)"
"by DNA methylation changes in promoters" was changed to "by DNA methylation changes in promoter proximity"
"regulated by promoter methylation" was changed to "regulated by promoter-proximal methylation"
"analysis of the promoter DMRs" was changed to "analysis of the promoter-proximal DMRs"
"between promoter methylation" was changed to "between promoter proximal methylation"
Cytoscape was used to analyse negatively or positively correlated DMR DEG pairs. ClueGO (v2.5.6) analysis was conducted using all DEG associated with a promoter proximal DMR (+/- 6 kb from TSS) and the Spearman correlation coefficient 0.5 (112).
Lines 299-301 - I'm not sure the graph in Fig S3A support the conclusion that there was a preferential negative relationship between DNAm and gene expression. Looks like there are a substantial number of cases where a positive relationship is observed and this needs to be acknowledged.
Response:
In this part, we refer to Fig S3C. In the left panel, downregulated genes clearly show higher counts for the hypermethylated DMRs, whereas the hypomethylated DMRs are enriched at upregulated genes (right panel), indicating a preference for negative correlation: lower methylation, higher gene expression. If there were no preference, we would expect a 50:50 ratio of hypo- and hypermethylated DMRs, and we observed a 77:23 ratio. Nevertheless, we agree that there is a substantial number of cases (n=151) with a high positive correlation, which we now highlight in the text. For clarity, we also modified the figure legend to indicate that a stacked histogram is represented in the panel.
__CHANGE IN THE MANUSCRIPT____: __
L303: Interestingly, 23.5% of the identified DMR DEG pairs (n=151) showed a positive correlation between gene expression and DNA methylation.
*Figure legend in Fig. S3C was changed to: C Stacked histogram showing location of hyper- and hypomethylated DMRs relative to the TSS of DEGs in downregulated (left) and upregulated (right) genes. *
Line 307 - what are the "analysed DEGs"? Are they the methylation associated genes?
Response:
Those are the DEGs we identified in RNA-seq analysis. To clarify, we changed the text to "identified DEGs".
__CHANGE IN THE MANUSCRIPT____: __
"analysed DEGs" was changed to "identified DEGs"*
Line 307-309 - "Among the analyzed DEGs, 76.5% (492) displayed a negative correlation (16.8% of the total DEGs), indicating a possible direct regulation by DNA methylation, while 23.5% (151) showed a positive correlation between gene expression and DNA methylation" - are the authors suggesting the positive correlation doesn't indicate direct regulation?
__Response: __
Thank you for highlighting this point. We did not intend to suggest that negative correlation indicates direct regulation, while positive correlation suggests a lack thereof. To clarify that point, we have reformulated this sentence.
__CHANGE IN THE MANUSCRIPT____: __
Among the identified DEGs, 76.5% (n=492) displayed a negative correlation (16.8% of the total DEGs), consistent with a repressive role of promoter DNA methylation. Interestingly, 23.5% of the identified DEG (n=151) showed a positive correlation between gene expression and DNA methylation.
Line 313 - why did the authors focus on only negatively correlated genes to identify their top dysregulated pathway of IFN signalling? Why not do pathway analysis on the DNAm associated genes separately to identify DNAm associated pathways?
Response:
We have also performed a pathway enrichment analysis using the positively correlated genes but did not identify any significantly enriched pathways/process/terms. When we examined the top hit of the gene set enrichment analysis, the interferon signaling pathway, we observed only negatively correlated DMR gene associations (Fig. 5B). Therefore, we decided to use only the negatively correlated DMRs, as using all correlated genes would give a higher background and dilute our results.
CHANGE IN THE MANUSCRIPT____:
Cytoscape was used to analyse negatively or positively correlated DMR DEG pairs. ClueGO (v2.5.6) analysis was conducted using all DEG associated with a promoter proximal DMR (+/- 6 kb from TSS) and the Spearman correlation coefficient 0.5 (113).
A comparison of the gene expression data with previous data in AT2 cell/single cell data would strengthen the gene expression section.
__Response: __
We compared our gene expression signatures with the study of Fujino et al., who profiled sorted AT2 cells (EpCAMhighPDPNlow) from COPD/controls using expression arrays (PMID: 23117565). Consistent with our study, the authors also observed the upregulation of interferon signalling (among other pathways) in COPD AT2s. However, no raw data was available in the published manuscript for a more in-depth analysis.
Several recent scRNA-seq studies identified transcriptional signatures of COPD and control cells (e.g., PMIDs: 36108172, 35078977, 36796082, 39147413__). However, most studies did not match the smoking status of the control and COPD donors and looked at the whole lung tissue, with limited power to detect gene expression changes in distal alveolar cells. It is difficult to directly compare our data to the gene expression data from non-smokers vs COPD patients, as cigarette smoking profoundly remodels the epigenome and transcriptional signatures of cells. In addition, differences in technologies and depth of sequencing make such comparisons challenging. However, one study (PMID: 36108172) performed scRNA-seq analysis on 3 non-smokers, 4 ex-smokers and 7 COPD ex-smoker lungs. Despite relatively limited coverage of epithelial cells in the dataset (We also compared the main AT2 IFN signature identified in the integration of our DNA methylation in promoter-proximal regions and RNA-seq with a recent study (published after the submission of our manuscript, PMID: 39147413) that profiled EpCAMpos cells from COPD and control lungs (non-smokers) using scRNA-seq. We observed an upregulation of our IFN signature genes in AT2 in COPD (specifically in AT2-c and rbAT2 subsets), suggesting that similar signatures were observed in this dataset as well. However, ex-smokers were not included in this study, making direct comparisons difficult. We have now included the panels shown below as __Figure S4E and S4F:
Figure S4E and F: Expression values for the indicated genes of the IFN pathway from an external scRNA-seq dataset of AT2 cells from COPD patients and healthy controls (74). Y-axis shows log-normalized gene expression levels. F. Combined gene set score of the genes shown in (E) in different subsets of AT2 cells from (74)*. The IFN signature genes were identified in our integrative analysis of TWGBS and RNA-seq in sorted AT2 cells. *
CHANGES IN THE MANUSCRIPT:
However, 5-AZA is a global demethylating agent, and the observed effects may not be direct. To validate the epigenetic regulation of central AT2 pathways further, we took advantage of locus-specific epigenetic editing technology (73). We focused on the IFN pathway because it was the most significantly enriched Gene Ontology (GO) term in our integrative analysis of TWGBS and RNA-seq data. Several IFN pathway members had associated hypomethylated DMRs within promoter-proximal regions and concomitant increased gene expression (Fig. 4C and Fig.S2C). Additionally, we confirmed the elevated expression of IFN-related genes with associated DMRs identified in our study in AT2 cells and AT2 cell subclusters from a recently published scRNA-seq cohort (74)* (Fig. S4E-F). *
(Methods) Validation of IFN gene upregulation in a published scRNA-seq dataset
scRNA-seq data from (74), generously provided by M. Köningshoff, were processed using the default Seurat workflow (117). Expression of IFN-related genes was extracted and plotted as log-normalised gene expression levels in AT2 cells from control and COPD donors. Seurat's AddModuleScore() function was used to compute a gene set score for a custom IFN program using the genes listed in __Fig. S4E __and to analyse the IFN gene set scores in AT2 cell subclusters identified in (74). Briefly, average gene expression scores were computed for the gene set of interest, and the expression of control features (randomly selected) was subtracted as described in (118).
Fig. S4 E and F. E. Expression values for the indicated genes of the IFN pathway from an external scRNA-seq dataset of AT2 cells from COPD patients and healthy controls (74). Y-axis shows log-normalized gene expression levels. F. Combined gene set score of the genes shown in (E) in different subsets of AT2 cells from (74). The IFN signature genes were identified in our integrative analysis of TWGBS and RNA-seq in sorted AT2 cells. __ __
The paragraph starting on line 173 feels a little redundant when we know there is RNA available to test if the differential DNAm links to altered gene expression - this selected of example regions/genes would be better placed after the gene expression has been reported, at which point you could say whether the linked genes displayed altered transcription.
Response:
The current structure (with DNA methylation, followed by RNA-seq and integration) is intentional and serves several important purposes. As this is the first genome-wide high-resolution COPD DNA methylation study of AT2, we aimed to describe the methylation landscape independently of gene expression (noting the limitation of current understanding of how DNA methylation regulates expression). This early focus on DMRs lays clear groundwork by highlighting potential regulatory elements and pathways that could be disrupted, independent of or even before corroborative transcriptional data. Additionally, positioning these examples early in the narrative helps to frame subsequent gene expression analyses. Once RNA data are introduced later, the reader can directly compare the methylation patterns with transcriptional outcomes, thereby enhancing the overall story. In other words, by first showcasing disease-relevant methylation changes, we underscore a hypothesis that these epigenetic modifications are functionally meaningful. The later integration of gene expression data then serves as a confirmatory or complementary layer, rather than the sole basis for inferring biological significance. This is important as we still do not fully understand the function of DNA methylation outside promoters, and its role is also important for splicing, 3D genome organisation, non-coding RNA regulation, enhancer regulation, etc.
Similarly, the TF enrichment analysis is great but maybe would have added value to be done on DNA regions later shown to be linked to differential expression - was there different enrichment at DNA regions that are vs are not associated with altered expression? And could you test in vitro whether changing methylation of DNA (maybe a blunt too like 5-aza would be ok) alters TF binding (cut+run/ChIP?). Furthermore, it would be interesting to understand the TF sensitivity analysis within the context of positive versus negative DNA methylation:gene expression correlations.
Response:
As suggested by the reviewer, we now performed the TF enrichment analysis using the DMRs with a high correlation (|correlation coefficient|>0.5) between methylation and expression (Figure S3D) and expanded the method section to include TF analysis. We observed ETS domain motifs enriched at hypomethylated regions. They prefer unmethylated DNA (MethylMinus) and are therefore expected to bind with higher affinity to the respective DMRs in COPD. We agree with the reviewer that further verifying altered TF binding using cut&run or ChIP assays would be very interesting, but it is out of the scope of this manuscript. Such analysis is technically very challenging to perform with low numbers of primary AT2 cells and will be the focus of our follow-up mechanistic studies.
CHANGE IN THE MANUSCRIPT____:
Additionally, motif analysis of DMRs that were highly correlated (|Spearman correlation coefficient| > 0.5) with DEGs revealed a prominent enrichment of the cognate motif for ETS family transcription factors, such as ELF5, SPIB, ELF1 and ELF2 at hypomethylated DMRs (Fig. S3D). Interestingly, SPIB was shown to facilitate the recruitment of IRF7, activating interferon signaling (71)*, and our WGBS data uncovers SPIB motifs at hypomethylated DMRs, which aligns with its binding preferences at unmethylated DNA (methyl minus, Fig. S3D). *
Figure S3D: Enrichment of methylation-sensitive binding motifs at hypo- (right) and hypermethylated (left) DMRs, using DMRs with a high correlation (|Spearman correlation coefficient| > 0.5) between methylation and gene expression. Methylation-sensitive motifs were derived from Yin et al (64). Transcription factors, whose binding affinity is impaired upon methylation of their DNA binding motif, are shown in red (Methyl Minus), and transcription factors, whose binding affinity upon CpG methylation is increased, are shown in blue (Methyl Plus).
(Methods) To obtain information about methylation-dependent binding for transcription factor motifs which are enriched at DMRs, the results of a recent SELEX study (64)* were integrated into the analysis. They categorised transcription factors based on the binding affinity of their corresponding DNA motif to methylated or unmethylated motifs. Those whose affinity was impaired by methylation were categorised as MethylMinus, while those whose affinity increased were categorised as MethylPlus. A motif database of 1,787 binding motifs with associated methylation dependency was constructed. The log odds detection threshold was calculated for the HOMER motif search as follows. Bases with a probability > 0.7 got a score of log(base probability/0.25); otherwise, the score was set to 0. The final threshold was calculated as the sum of the scores of all bases in the motif. Motif enrichment analysis was carried out against a sampled background of 50,000 random regions with matching GC content using the findMotifsGenome.pl script of the HOMER software suite, omitting CG correction and setting the generated SELEX motifs as the motif database. *
__Methods: __ • The authors should include more detail of the TWGBS rather than directing the reader to a previous publication. Also DNA concentration post bisulfite conversion would be a useful metric to provide.
__Response: __
Following the suggestion, we have now expanded the details of TWGBS in the methods part of the manuscript. Due to limited space, we did not include a detailed protocol but instead referred to a published step-by-step protocol (55). Of note, we do not measure DNA concentration post-bisulfite conversion but consistently use the starting input of 30 ng of genomic DNA across all samples.
__CHANGE IN THE MANUSCRIPT____: __
(Methods): 15 pg of unmethylated DNA phage lambda was spiked in as a control for bisulfite conversion. Tagmentation was performed in TAPS buffer using an in-house purified Tn5 assembled with load adapter oligos (55) at 55 {degree sign}C for 8 min. Tagmentation was followed by purification using AMPure beads, oligo replacement and gap repair as described (55). Bisulfite treatment was performed using EZ DNA Methylation kit (Zymo) following the manufacturer's protocol.
*The T-WGBS library preparations were performed for all donors in parallel and sequenced in a single batch to minimize batch effects and technical variability. *
Differential DNA methylation analysis: It is stated that DNA regions had to contain 3 CpG sites but was this within a defined DNA size range?
Response:
The maximum distance between individual CpGs within DMR was set to 300 bp. To clarify, we added that information to the methods part.
__CHANGE IN THE MANUSCRIPT____: __
*"regions with at least 10% methylation difference and containing at least 3 CpGs with a maximum distance of 300 bp between them. *
Refence genome only provided for RNAseq not TWGBS?
__Response: __We used hg19 as the reference genome. The information on the reference genome for DNA methylation analysis was provided in the methods L574 (original manuscript_: "The reads were aligned to the transformed strands of the hg19 reference genome using BWA MEM")
The tables do not appear in the PDF and I struggled to tally to the "Dataset" files provided if that is what they were referring to?
Response:
Full tables (uploaded as Datasets in the manuscript central due to their size) were uploaded together with the manuscript files. They are quite large and will not convert to pdf, so they may not have been included in the merged pdf file. We assume that they should be available to the reviewers with the other files and will clarify that with the editorial staff in the resubmission cover letter.
For the gene expression analysis, can it be made clearer that a full analysis was done on COPD I samples. It is a little confusing to the reader as this was not done for DNAm so might be assumed the same targeted analysis on only genes found to be differentially expressed between control and COPD II-IV, but that cannot be the case as an overlap of COPD1 vs COPD II-IV genes if provided. For this overlap, do genes show the same effect direction?
__Response: __
To clarify, for the RNA-seq analysis, we performed DEG analysis for no-COPD versus COPD II-IV, as well as no-COPD versus COPD I. We then took all differentially expressed genes (presented in the Venn diagram) and plotted them for all samples as a heatmap. To split the genes into groups displaying similar effect directions, we applied a clustering approach and identified 3 main signatures. Cluster 3 primarily comprises genes unique to COPD I samples, which are associated with the adaptive immune system and hemostasis (Fig. 4E). In the other two clusters, we mainly observe a transitioning pattern from control to severe COPD samples, correlating with the FEV1 values of the patients. This has now been clarified in the manuscript.
Replication is difficult on these studies as the samples are so difficult to come by. Also limited by sample size for the same reason. It doesn't mean the study is not worth doing and the data are still valuable. However, it may be pertinent to include technical validation of a few regions of interest, acknowledge the limitation (along side strengths) in the discussion, and perhaps provide actual p value rather than blanket Response:
We thank the reviewer for acknowledging the replication challenges for studies working with sparse human material and hard-to-purify cell populations. Following the reviewer's suggestion, we have now included a strengths and limitations section in the discussion where we summarised the points highlighted by both reviewers.
Regarding technical validation, we would like to note that the whole genome bisulfite sequencing (WGBS) technology, as well as the tagmentation-based WGBS (T-WGBS), have been validated in the past few years in several publications (e.g., PMID: 24071908) and shown to yield reliable DNA methylation quantification in comparison to other technologies (PMID: 27347756). For us, technical validation using alternative methods (e.g. bisulfite sequencing or pyrosequencing) is difficult as it requires significantly more input DNA than the low-input T-WGBS we have performed and obtaining sufficient amounts of material from primary human AT2 cells (especially from severe COPD) is not possible with the size of tissue we can access. However, while establishing the T-WGBS for this project, we initially validated our approach using Mass Array, a sequencing-independent method. For this, we performed T-WGBS on the commercially available smoker and COPD lung fibroblasts and selected 9 regions with different methylation levels for validation using a Mass Array. We obtained an excellent correlation between both methods, providing technical validation of T-WGBS and our analysis workflow. This validation was published in our earlier manuscript (PMID: 37143403), but we provided the data below for convenience.
Scatter plots showing correlation of average methylation obtained with T-WGBS and Mass Array from COPD and smoker fibroblasts. Each dot represents one region with varying methylation levels. The blue diagonal represents the linear regression. Shaded areas are confidence intervals of the correlation coefficient at 95%. Correlation coefficients and P values were calculated by the Pearson correlation method.
To enable further validation and follow-up by the community, we included the full list of DMRs, associated p-values and additional information for DNA methylation analysis (DMR width, n.CpGs, MethylDiff, etc) in Table 3 (Table_3_wgbs_dmr_info.xlsx) and the information about DEGs from RNA-seq in Table 6 (Table_6_RNAseq_DEG_info.xlsx).
It isn't clear to me if DNA and RNA are from the same cells? The results say "cells matching those used for T-WGBS" but the methods suggest separate extractions so not the same cells? If they are not the same cells a comment on the implications of this should be included in the discussion for example, potentially some differences in cell type composition, storage time etc.
Response:
Lung tissue samples were freshly cryopreserved, and H&E slides derived from exemplary pieces of the tissue analyzed. Once we had a group of at least 3 samples comprising one non-COPD and 2 COPD samples, we processed them in parallel to limit sorting variation between control and disease samples. The sorted cells were counted, aliquoted and pelleted at 4{degree sign}C before flash freezing and storing at -80{degree sign}C. The storage time of the cell pellets varied between the donors. RNA and DNA were isolated from cell pellets collected from the same FACS sorting experiment; therefore, we do not expect differences in cell type composition. In addition, RNA and DNA isolation were performed for all sorted pellets in parallel. All library preparations for TWGBS and RNA-seq were performed for all donors in parallel and sequenced in a single batch to minimise batch effects and technical variability. This has now been clarified in the methods part of the manuscript.
__CHANGE IN THE MANUSCRIPT____: __
To minimize potential technical bias, samples from no COPD and COPD donors were processed in parallel in groups of 3 (one no COPD and 2 COPD samples).
RNA and genomic DNA for RNA-seq and TWGBS were isolated from identical aliquots of sorted cell pellets.
Genomic DNA was extracted from 1-2x104 sorted alveolar epithelial cells isolated from cryopreserved lung parenchyma from 11 different donors in parallel using QIAamp Micro Kit
The TWGBS library preparations were performed for all donors in parallel and sequenced in a single batch to minimize batch effects and technical variability.* *
RNA was isolated from flash-frozen pellets of 2x104 sorted AT2 cells from 11 different donors in parallel.
The RNA-seq library preparation for all donors was performed in parallel and all samples were sequenced in a single batch to minimize batch effects and technical variability.
Line 193 the authors say "Since DMRs were overrepresented at cis-regulatory sites...." - "cis" needs to be defined. If you link DNAm regions to gene via "closest gene" does this not automatically mean you're outputs will be cis? Just needs better definition/explanation.
Response:
The term "cis‐regulatory sites" in our manuscript is intended to denote regulatory elements-such as enhancers, promoters, and other nearby control regions-that reside on the same chromosome and close to the genes they regulate. While it's true that linking a DMR to its closest gene captures a cis association, our phrasing emphasises that the DMRs are enriched specifically at these functional regulatory elements (Fig. 2E) rather than being randomly distributed. This usage aligns with established conventions in the field. To avoid any misunderstandings, we have now changed the term to gene regulatory sites.
__CHANGE IN THE MANUSCRIPT____: __
*We changed the "cis-regulatory sites" to "gene regulatory sites" *
__Minor comments: __
Line 157: "we identified site-specific differences....". Change to region specific?
Response:
This has now been corrected as suggested.
Line 102-103: needs a reference for the statement "Alterations in DNA methylation patterns have been implicated......"
Response:
Following the reviewer's suggestion, we added the relevant references (34-36) to this statement.
Line 266 - what does "strong dysregulation" mean? Large fold change, very significant?
Response:
We removed the word "strong" from this sentence.
Lines 423-425 - statement needs a reference
Response:
Following the reviewer's suggestion, we added the relevant reference to this statement.
Line 428 - word missing between "epigenetic , we"?
Response:
This has now been corrected. The text reads: "Through treatment with a demethylating drug and targeted epigenetic editing, we demonstrated the ability to modulate..."
Prior studies are well references, text and figures are clear and accurate.
__Reviewer #2 (Significance (Required)): __
This study has several strengths:
1) Sample collection and characterisation. AT2 cells are incredibly hard to come by and the authors should be commended to generating the samples. However, proximity to cancer is always a potential issue, especially in epigenetic studies. Is it feasible to include any analysis to show the samples derived from those with cancer don't drive the changes observed? Even a high level PCA or an edit of fig 2A with non-cancer in a different colour in supplemental - looks like there is one outlier, is that a non-cancer? Or a correlation of change in beta between control and cancer/COPD and control and non-cancer:COPD (for want a better phrase!). just an indicator that the non-cancer COPD samples are not driving differences.
Response:
We thank the reviewer for highlighting the value of generating data from hard-to-work-with AT2 populations and bringing up the important point of cancer proximity, which we considered very carefully when designing our study. To match our samples across the cohort, all the no-COPD, COPD I, and two of the COPD II-IV distal lung samples were obtained from cancer resections. In addition to other characteristics, like age, BMI and smoking status, we also matched the donors by cancer type (all profiled donors had squamous cell carcinoma). We collected lung tissue as far away from the carcinoma as possible and sent representative pieces for histological analysis by an experienced lung pathologist to confirm the absence of visible tumours. In addition, to ensure that our data represents COPD-relevant signatures, we intentionally included samples from three COPD donors undergoing lung resections (without a cancer background) in the profiling.
Following the reviewer's suggestion, to investigate the potential impact of non-cancer samples on driving the observed differences, we carefully checked the PCAs for both DNA methylation and RNA-seq. We could not identify a clear separation of no-cancer COPD samples from the cancer COPD samples (or other cancer samples) in any examined PCs, indicating no cofounding effect of cancer samples. We observed that one sample contributing to PC2 is a non-cancer sample, but this was a rather sample-specific effect, as the other two non-cancer samples clustered together with the other severe COPD samples with a cancer background. Notably, in our DNA methylation data, we do not observe typical features of cancer methylomes, like global loss of DNA methylation or aberrant methylation of CpG islands (e.g., in tumour suppressor genes) (see Fig. 2A), further suggesting that we do not "pick up" confounding cancer signatures in our data.
Following the comments from both reviewers, to clarify that point, we added the information about cancer and non-cancer samples to the PCA figures for DNA methylation (new Fig. 2B) and RNA-seq (new Fig. 3A) data in the revised manuscript, as shown below
CHANGE IN THE MANUSCRIPT____:
COPD samples from donors with a cancer background clustered together with the COPD samples from lung resections, confirming that we detected COPD-relevant signatures (Fig. 2B).
Fig. 2B.* Principal component analysis (PCA) of methylation levels at CpG sites with > 4-fold coverage in all samples. COPD I and COPD II-IV samples are represented in light and dark green triangles, respectively, and no COPD samples as blue circles. COPD samples without a cancer background are displayed with a black contour. The percentage indicates the proportion of variance explained by each component. *
Unsupervised principal component analysis (PCA) on the top 500 variable genes revealed a clear influence of the COPD phenotype in separating no COPD and COPD II-IV samples, as previously observed with the DNA methylation analysis, irrespective of the cancer background of COPD samples (Fig.3A, Fig. S2B).
*Principal component analysis (PCA) of 500 most variable genes in RNA-seq analysis. PCA 1 and 2 are shown in Fig.3A, PCA 1 and 4 in Fig.S2B. COPD I and COPD II-IV samples are represented in light and dark green triangles, respectively, and no COPD samples as blue circles. COPD samples without a cancer background are displayed with a black contour. The percentage indicates the proportion of variance explained by each component. *
2) This is the first time DNAm has been profiled in AT2 cells. It is incredibly difficult, valuable and novel data that will increase the fields capability technically, their understanding of functional mechanisms and potential translation considerably. It's audience will be primarily translational respiratory however the fundamental science aspect of gene expression regulation by DNA methylation with have wider reach across developmental and disease science.
Response:
We thank the reviewer for recognising the uniqueness and novelty of our study and highlighting the value and potential impact of our datasets for the lung field.
3) the functional analysis using targeted CRISPR-Cas9 is very well done and adds impact.
Response:
We thank the reviewer for recognising the strengths and added value of the functional analysis using epigenetic editing.
__Potential weaknesses/areas for development __
I feel the main weakness is the in the section integrating DNA methylation and gene expression. The rationale for a focus on various aspects, for example inversely related DNAm/gene expression pairs, the IFN pathway and IRF9, are not clear. Also further understanding of the differences between DNAm associated genes and non-DNAm associated genes could be expanded, at the pathway level, TF regulation level, effect size level (are DNAm associated changes to gene expression larger, enriched for earlier differential expression)
Response:
Our rationale for focusing on the inversely related DNAm/gene expression pairs in promoter proximal is purely data-driven, as they represent the biggest group in our data (Fig. 4A-B). Among those negatively correlated genes, we observed the strongest enrichment for the IFN pathway (Fig. C), making it an obvious, data-driven target for further studies. The negative correlation of expression and methylation for IFN pathway genes could be validated in 5-AZA assays in A549 cells (Fig. 5A). Next, we made an interaction network analysis showing IRF9 and STAT2 as master regulators (Fig. 5B) of the negatively correlated IFN genes. As IRF9 itself displayed a negative correlation between DNA methylation and expression (Fig. 5C), we used the associated DMR for further epigenetic editing (Fig. 5D-E). We performed the additional requested analyses of the enhancer-associated changes and genes, as described above. We fully agree with the reviewer that our data sets are a great resource and can be further used to elaborate on other relationships of DNA methylation and RNA expression or other pathways, but this is out of the scope of this study. To enable further studies by the research community, we provide all necessary information about DMRs and DEGs in the associated supplementary tables and the raw data through the EGA, as well as the CRISPRa editing assay.
The authors could comment on potential masking of differences between 5hmC and mC and the implications it may have
Response:
We thank the reviewer for bringing up this important point. Indeed, bisulfite sequencing cannot differentiate between methylated and hydroxymethylated cytosines; hence, some of the methylated sites may be hydroxymethylated. However, the overall levels of hydromethylation in differentiated adult tissues are very low (except for the brain), orders of magnitude lower compared to DNA methylation. Following the reviewer's suggestion, we have added a sentence in the limitation section of the discussion to clarify that point.
__CHANGE IN THE MANUSCRIPT: __
In addition, while WGBS provides unprecedented resolution and high coverage of the DNA methylation sites across the genome, it does not allow distinguishing 5-methylcytosine from 5-hydroxymethylcytosine. Therefore, we cannot exclude that some methylated sites we detected are 5-hydroxymethylated. However, the 5-hydroxymethylcytosine is present at very low levels in the lung tissue (97)*. ** *
Furthermore, while the rationale for looking at DMRs is clear, especially given the sample number, I am interested to understand what proportion of the assayed CpGs "fit" within the cut off stipulations of the DMR analysis - that is, is their potentially COPD effects at sparse CpG regions/individual CpG sites that are not being identified. A comment on this would be useful and seems the strength of profiling genome wide. I'm happy genome wide is beneficial it just feels a little circular that the authors have chosen whole genome to avoid the bias of the Illumina array and a focus on promotors, but have primarily reported promoter DNAm. This caught my attention again in the discussion where the authors state that cis-regulatory regions were also identified in their fibroblast data .....is this finding a factor of the analysis performed? (also a comparison of regions Identified in AT2 cells versus fibroblasts would be really interesting for a future paper)
Response:
We decided to focus our analysis on regions rather than individual CpG sites when looking at differential methylation, as DNA methylation is spatially correlated, and methylation changes in larger regions are more likely to have a biological function. Extending the analysis to single CpG sites would require a higher number of samples for a reliable analysis compared to the DMR analysis (as mentioned by the reviewer).
Of note, we addressed the platform comparison between Illumina array technology and WGBS in our previous fibroblast study (PMID: 37143403), where we compared our WGBS data with the published 450k array data of COPD parenchymal fibroblasts (Clifford et al., 2018). We observed only a marginal overlap between the CpGs from our DMRs and the CpGs probes available on the array (which was due to the differences in technologies used and the limited coverage of the 450K array in comparison to our genome-wide approach, in which we covered 18 million CpGs). Out of the 6279 DMRs identified in our fibroblast study, only 1509 DMRs overlapped with at least one CpG probe on the 450K array, and after removing low-quality CpGs from the array data, only 1419 DMRs were left. This comparison highlighted the increased resolution of the WGBS compared to Illumina arrays.
The reason why we focused on promoter proximal DMRs are the following: 1) the assignment of the enhancer elements in AT2 to the corresponding gene is still too inaccurate in the absence of AT2 specific enhancer chromatin maps 2) regulation at enhancers by DNA methylation might be more complex and might change (increase or attenuate) binding affinities of certain transcription factors (Fig.2H), which might lead to gene expression changes or 3) methylation changes might be an indirect effect of differential TF binding PMID: 22170606). However, we agree with the reviewer that despite these limitations, expanding the analysis beyond promoters adds value to the manuscript; hence, as described above, we expanded the analysis of non-promoter regions, including enhancers, in the revised manuscript.
We thank the reviewer for the suggestion to compare the regions identified in AT2 cells and fibroblasts in a future paper.
My expertise:Respiratory, cell biology, epigenetics.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
This study aim to understand the molecular mechanisms underlying dysfunction in AT2 cells in COPD, by profiling bulk genome wide DNA methylation using Tagmentation-based whole-genome bisulfite sequencing (T-WGBS) and RNA sequencing in selectively sorted primary AT2 cells. The study stands out in it's sequencing breadth and use of an incredibly difficult cell population, and has the potential to add substantially to our mechanistic understanding of epigenetic contributions to COPD. A further highlight is the concluding aspect of the study where the authors undertook targeted modification of specific CpG methylation, provided direct, site-specific evidence for transcriptional regulation by CpG methylation.
Major comments:
The authors clearly show that there is DNA methylation alteration in AT2 cells from COPD individuals that links functional to gene expression at some level. However, I think the statement "to identify genome-wide changes associated with COPD development and progression..." and similar other references to disease development understanding is not accurate given the DNA methylation primary comparison is between control and moderate to severe COPD, with no temporal detail or evidence that they drive progression rather than are a result of COPD development. The paragraph starting on line 186 where this is a addressed to some extent is quite vague and doesn't really provide confidence that DNAm dysregulation occurs at an early stage in this context. This can be addressed by changing the focus/style of the text.
Results comments and suggestions:
For the integrated analysis, there is a focus on DMRs in promoters with very little analysis on other regions. The paragraph starting on line 317 describes some analysis on enhancers but is very brief, doesn't include information on how many/which DMRs were included, making it hard to interpret the impact of the 147 DMRs and 93 genes identified - is this nearly all DMRs and genes analysed or very few? A comparison to the promoter analysis would be of interest. Especially as the targeted region followed up with lovely functional assessment in the last sections is a gene body DMR, not a promoter DMR.
Lines 299-301 - I'm not sure the graph in Fig S3A support the conclusion that there was a preferential negative relationship between DNAm and gene expression. Looks like there are a substantial number of cases where a positive relationship is observed and this needs to be acknowledged.
Line 307 - what are the "analysed DEGs"? Are they the methylation associated genes?
Line 307-309 - "Among the analyzed DEGs, 76.5% (492) displayed a negative correlation (16.8% of the total DEGs), indicating a possible direct regulation by DNA methylation, while 23.5% (151) showed a positive correlation between gene expression and DNA methylation" - are the authors suggesting the positive correlation doesn't indicate direct regulation?
Line 313 - why did the authors focus on only negatively correlated genes to identify their top dysregulated pathway of IFN signalling? Why not do pathway analysis on the DNAm associated genes separately to identify DNAm associated pathways?
A comparison of the gene expression data with previous data in AT2 cell/single cell data would strengthen the gene expression section.
The paragraph starting on line 173 feels a little redundant when we know there is RNA available to test if the differential DNAm links to altered gene expression - this selected of example regions/genes would be better placed after the gene expression has been reported, at which point you could say whether the linked genes displayed altered transcription.
Similarly, the TF enrichment analysis is great but maybe would have added value to be done on DNA regions later shown to be linked to differential expression - was there different enrichment at DNA regions that are vs are not associated with altered expression? And could you test in vitro whether changing methylation of DNA (maybe a blunt too like 5-aza would be ok) alters TF binding (cut+run/ChIP?). Furthermore it would be interesting to understand the TF sensitivity analysis within the context of positive versus negative DNA methylation:gene expression correlations.
Methods:
The authors should include more detail of the TWGBS rather than directing the reader to a previous publication. Also DNA concentration post bisuphite conversion would be a useful metric to provide.
Differential DNA methylation analysis: It is stated that DNA regions had to contain 3 CpG sites but was this within a defined DNA size range?
Refence genome only provided for RNAseq not TWGBS?
The tables do not appear in the PDF and I struggled to tally to the "Dataset" files provided if that is what they were referring to?
For the gene expression analysis, can it be made clearer that a full analysis was done on COPD I samples. It is a little confusing to the reader as this was not done for DNAm so might be assumed the same targeted analysis on only genes found to be differentially expressed between control and COPD II-IV, but that cannot be the case as an overlap of COPD1 vs COPD II-IV genes if provided. For this overlap, do genes show the same effect direction?
Replication is difficult on these studies as the samples are so difficult to come by. Also limited by sample size for the same reason. It doesn't mean the study is not worth doing and the data are still valuable. However, it may be pertinent to include technical validation of a few regions of interest, acknowledge the limitation (along side strengths) in the discussion, and perhaps provide actual p value rather than blanket < p 0.1, seems very lenient but may all be super significant (this may already be in the tables I wasn't able to find).
It isn't clear to me if DNA and RNA are from the same cells? The results say "cells matching those used for T-WGBS" but the methods suggest separate extractions so not the same cells? If they are not the same cells a comment on the implications of this should be included in the discussion for example, potentially some differences in cell type composition, storage time etc.
Line 193 the authors say "Since DMRs were overrepresented at cis-regulatory sites...." - "cis" needs to be defined. If you link DNAm regions to gene via "closest gene" does this not automatically mean you're outputs will be cis? Just needs better definition/explanation.
Minor comments:
Line 157: "we identified site-specific differences....". Change to region specific?
Line 102-103: needs a reference for the statement "Alterations in DNA methylation patterns have been implicated......"
Line 266 - what does "strong dysregulation" mean? Large fold change, very significant?
Lines 423-425 - statement needs a reference
Line 428 - word missing between "epigenetic , we"?
Prior studies are well references, text and figures are clear and accurate.
This study has several strengths:
1) Sample collection and characterisation. AT2 cells are incredibly hard to come by and the authors should be commended to generating the samples. However, proximity to cancer is always a potential issue, especially in epigenetic studies. Is it feasible to include any analysis to show the samples derived from those with cancer don't drive the changes observed? Even a high level PCA or an edit of fig 2A with non-cancer in a different colour in supplemental - looks like there is one outlier, is that a non-cancer? Or a correlation of change in beta between control and cancer/COPD and control and non-cancer:COPD (for want a better phrase!). just an indicator that the non-cancer COPD samples are not driving differences.
2) This is the first time DNAm has been profiled in AT2 cells. It is incredibly difficult, valuable and novel data that will increase the fields capability technically, their understanding of functional mechanisms and potential translation considerably. It's audience will be primarily translational respiratory however the fundamental science aspect of gene expression regulation by DNA methylation with have wider reach across developmental and disease science.
3) the functional analysis using targeted CRISPR-Cas9 is very well done and adds impact.
Potential weaknesses/areas for development:
I feel the main weakness is the in the section integrating DNA methylation and gene expression. The rationale for a focus on various aspects, for example inversely related DNAm/gene expression pairs, the IFN pathway and IRF9, are not clear. Also further understanding of the differences between DNAm associated genes and non-DNAm associated genes could be expanded, at the pathway level, TF regulation level, effect size level (are DNAm associated changes to gene expression larger, enriched for earlier differential expression) The authors could comment on potential masking of differences between 5hmC and mC and the implications it may have
Furthermore, while the rationale for looking at DMRs is clear, especially given the sample number, I am interested to understand what proportion of the assayed CpGs "fit" within the cut off stipulations of the DMR analysis - that is, is their potentially COPD effects at sparse CpG regions/individual CpG sites that are not being identified. A comment on this would be useful and seems the strength of profiling genome wide. I'm happy genomewide is beneficial it just feels a little circular that the authors have chosen whole genome to avoid the bias of the Illumina array and a focus on promotors, but have primarily reported promoter DNAm. This caught my attention again in the discussion where the authors state that cis-regulatory regions were also identified in their fibroblast data ..... is this finding a factor of the analysis performed? (also a comparison of regions Id'ed in AT2 cells versus fibroblasts would be really interesting for a future paper)
My expertise: Respiratory, cell biology, epigenetics.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
This study by Prada et al. aimed to explore DNA methylation and gene expression in primary EpCAMhigh/PDPNlow cells, consisting for (probably) the largest part of AT2 cells, to understand the molecular mechanisms behind the impaired regeneration of alveolar epithelial progenitor cells in COPD. They found that higher or lower promoter methylation in COPD-associated cells was inversely correlated with changes in gene expression, with interferon signaling emerging as one of the most upregulated pathways in COPD. IRF9 was identified as the master regulator of interferon signaling in COPD. Targeted DNA demethylation of IRF9 in an A549 cell line resulted in a robust activation of its downstream target genes, including OAS1, OAS3, PSMB8, PSMB9, MX2 and IRF7, demonstrating that demethylation of IRF9 is sufficient to activate the IFN signaling pathway, validating IRF9 as a master regulator of IFN signaling in (alveolar) epithelial cells.
Major comments:
To remove airways (and blood vessels) completely from the lung tissue is difficult, if not impossible. This means that the assumption that the sorted EpCAMpos/PDPNlow cells primarily consisted of AT2 cells remains valid only if a quantitative analysis is conducted on the proportion of HT2-280pos cells in all samples in cytospins to exclude any significant contamination from bronchial epithelial cells. If authors cannot demonstrate >95% pure HT-280-positive cells, then the key conclusions suggesting that the epigenetic regulation of the IFN pathway might be crucial in AT2 progenitor cell regeneration could also potentially apply to bronchial progenitor cells. In addition, if >95% purity can not be demonstrated, the data should be adjusted to account for differences in cell type composition.
The overrepresentation of several keratins (KRT5, KRT14, KRT16, KRT17), mucins (MUC12, MUC13, MUC16, MUC20) and the transcription factor FoxJ1 is now attributed by the authors to a possible dysregulation of AT2 identity and differentiation in COPD (lines 282 - 284) where they cite refs 28, 69, 70. Authors try to support this with IF double stains for KRT5 and HT-280 to identify co-expression of KRT5 and HT2-280 in lung tissue (Figure S2H). However, the evidence for the co-expression of both markers could be presented more convincingly.
Double staining for KRT5 and HT2-280 did highlight the proximity of both cell types in lung tissue, underscoring the challenge of removing airways (including the smaller and terminal bronchi) from the tissue. In addition, HT-280/KRT5 co-expression in not consistent with recent studies from refs 28, 69, 70 where other markers for distal airway cell transition, such as SCGB3A2 and BPIFB1, have been demonstrated, which were not investigated in this study.
The small (and not evenly divided) sample size of both COPD and non-COPD specimens may lead to a higher risk for false positive results as adjustments for multiple testing typically rely on the number of comparisons, and small sample sizes may not provide enough data points to adequately control for this.
Minor comments:
Introduction:
In general, refer to the actual experimental studies rather than review papers where appropriate.
Clearly specify whether a study was conducted in mice or humans, as this distinction is crucial for understanding the relevance of the findings to COPD.
Methods:
Discussion:
A list of limitation should be added to the discussion. One is the use of the alveolar cell line A549, which produces mucus, a characteristic more commonly associated with bronchial epithelial cells. (ref 43)
Another limitation to consider is that cells were isolated primarily from individuals with lung cancer, except for patients with COPD stage IV. In particular as COPD stage II and IV samples were taken together.
And discuss the small and unevenly divided sample size
References:
Check references. For instance, there is no reference in the text to ref 43.
Align format of references
The strength of this study lies in its focus on the molecular mechanisms underlying the impaired regeneration of epithelial progenitor cells in COPD. The discovery of IRF9, which regulates IFN signaling and is prominently upregulated in COPD, together with the convincing validation of the epigenetic control of the IFN pathway by targeted DNA demethylation of the IRF9 gene, adds significant value to the COPD research field.
Main limitations of the study are the relatively small sample size of both COPD and non-COPD specimens and the claim that the sorted EpCAMpos/PDPNlow cells primarily consisted of AT2 cells.
The nature and significance of the advance in epigenetic editing of IRF9 in COPD can be described as both conceptual and potentially clinical: Conceptual Advance: The epigenetic editing of IRF9 enhances our understanding of the molecular mechanisms underlying COPD pathogenesis. By targeting IRF9 through epigenetic modifications, researchers were able to modulate the activity of the IFN pathway, which plays a crucial role in the immune response and lung tissue regeneration. This approach offers insights into the epigenetic regulation of gene expression in epithelial progenitor cells in COPD and expands our understanding of how alterations in specific gene methylation could contribute to disease progression. Clinical Significance: The potential clinical significance of epigenetic editing of IRF9 lies in its implications for COPD therapy. If successful, epigenetic editing techniques could offer a novel therapeutic strategy for COPD by downregulating IFN pathway activation and promoting regeneration of epithelial progenitor cells in the lungs. Obviously, further preclinical and clinical studies are needed to validate the efficacy and safety of epigenetic editing approaches in COPD treatment.
Few experimental papers have been published on epigenetic editing in lung diseases, with limited research available beyond the study referenced in citation 43. Song J, Cano-Rodriquez D, Winkle M, Gjaltema RA, Goubert D, Jurkowski TP, Heijink IH, Rots MG, Hylkema MN. Targeted epigenetic editing of SPDEF reduces mucus production in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2017 Mar 1;312(3):L334-L347. doi: 10.1152/ajplung.00059.2016. Epub 2016 Dec 23. PMID: 28011616.
This study is of broad interest to researchers investigating the pathogenesis and treatment of COPD.
Expertise in: Lung pathology, Immunology, COPD, Epigenetics
Less expertise in: Epigenetic Editing
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
Summary: findings and key conclusions Epithelial cell competition in larval imaginal discs involves signaling with the Sas ligand and Ptd10D receptor. In wild type cells both are typically found at the apical surface, but relocalize to the lateral cortex at the winner-loser interface. Ptd10D activation leads to reduced Ras signaling, increased pro-apoptotic Jnk signaling and consequently the elimination of loser cells. In the manuscript the authors address the role of the actin cytoskeleton in the context of the signaling controlling cell elimination in Drosophila larval eye imaginal discs. They interfere by clonal overexpression of the guanyl nucleotide exchange factor RhoGEF2 (RG2), which has previously been shown to induce dominant gain-of-function phenotypes by activation of Rho signaling. In this context the requirement of and genetic interactions with the other pathways implicated in cell elimination is tested. They find that RG2 induced cell elimination depends on PtD10D, Hippo signaling and Crumbs.
Major comments: claims and conclusions The experimental setting, using clonal analysis in imaginal discs, is straight-forward and well-established, including quantification of clone size and comparison of phenotypes. The presented data are of high quality and thus the direct conclusions are fully supported by the data as long as they refer to the actual experimental interference. What is not supported by the data is the generalization of the conclusions, i. e. that RG2 overexpression would be equivalent to Actin cytoskeletal deregulation. This equivalence is expressed in the title "Actin cytoskeletal deregulation, caused by RhoGEF2 overexpression.." and the summary " that actin cytoskeleton deregulated cells (as induced by RhoGEF2 overexpression (RhoGEF2OE))...". In my view such an equivalence is not justified. There is no doubt that RG2 overactivation affects the actin cytoskeleton in multiple ways, such as contractility via MyoII or polymerization via Dia, among others. There is also no doubt that other pathways are also directly or indirectly affected beside the actin cytoskeleton. The authors do not present data showing the specificity of RG2 overexpression. For example, the authors could investigate the phenotype and genetic interaction with an alternative way of interference, independent of RG2, of the actin cytoskeleton to support their conclusion. There is a second assumption, which may not be justified, that the function of the cytoskeleton would be generally downstream of cell polarity, see abstract l24 "triggering cytoskeletal deregulation (which occurs downstream of cell polarity disruptions)..". There are certainly cytoskeletal activities such as cell shape changes that mediate the execution of cell elimination. However interfering with the cortical cytoskeleton also affect the distribution of cortical polarity proteins. The authors do not present data to demonstrate the specificity of RG2 overexpression concerning a function downstream of cell polarity.
Response: We apologise for our phrasing of the title and the sentence in the summary that suggests that it is the actin cytoskeleton disruption caused by RhoGEF2 overexpression that is responsible for the effects on cell competition. We have rephrased the title and edited the text to avoid such an inference.
With regard to the reviewer’s second concern regarding the link between cell polarity disruption and actin cytoskeletal deregulation, there is indeed evidence that this occurs.
There are numerous examples of how cell polarity regulators affect the actin cytoskeleton in both Drosophila and mammalian cells (reviewed by Humbert et al., 2015, DOI 10.1007/978-3-319-14463-4_4). Indeed, in our previous paper (Brumby et al., 2011. PMID: 21368274), we found genetic evidence that the knockdown of the polarity regulator, dlg, cooperates with activated Ras (RasACT) to produce a hyperplastic eye phenotype, and that this phenotype is rescued by knockdown of actin cytoskeletal regulators like RhoGEF2 or Rho. This data suggests that these actin cytoskeleton regulators act downstream of cell polarity disruption to cooperate with RasACT. Furthermore, another study has shown that the activation of Myosin II is increased in scrib mutants and impairs Hippo pathway signaling, and is also required for the cooperation of scrib mutants with RasACT (Külshammer, et al., 2013. PMID: 23239028). Consistent with this finding, we have previously shown that RhoGEF2 acts via Rho, Rok, and Myosin II activation in cooperation with RasACT (Khoo et al., 2013. PMID: 23324326). Furthermore, another cell polarity regulator, Lgl, binds to and negatively regulates Myosin II function in Drosophila (Strand et al., 1994. PMID: 7962095; Betschinger et al., 2005. PMID: 15694314). Moreover, Drosophila Scrib and Dlg bind to GUK-holder/NHS1 (Nance–Horan syndrome-like 1), which is a regulator of the WAVE/SCAR-ARP2/3-branched F-actin pathway, and this interaction is required for epithelial tissue development (Caria et al., 2018. PMID: 29378849). Thus, although cell polarity gene loss can affect the actin cytoskeleton by different means, and RhoGEF2 can activate Rho to regulate various actin cytoskeletal effectors (Limk, Dia, PKN, Rok), what they have in common is the activation of Myosin II. To make this clearer, we have now added brief sections to the introduction and Discussion highlighting and contextualising evidence for the effect of cell polarity disruption on the actin cytoskeleton.
Reviewer #1 (Significance (Required)):
The study establishes genetic interactions and dependencies concerning cell elimination following a very specific experimental interference of RG2 overexpression. It remains unclear, however, to which degree these genetic interactions contribute to controlling cell competition in situations that are physiologically relevant. The generalization of RG2 overexpression as a specific test the function of the actin cytoskeleton is an interpretation not supported by the presented data and the experimental set up.
Response: Although RhoGEF2 overexpression does lead to actin cytoskeletal disruption via Rho effectors, the reviewer is correct that we do not know whether it is the actin cytoskeleton disruption per se that is involved in triggering cell competition. We have edited the text accordingly.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
Summary: In the manuscript "Actin cytoskeletal deregulation, caused by RhoGEF2 overexpression, induces cell competition dependent on Ptp10D, Crumbs, and the Hippo signaling pathway", Natasha et al. investigate how actin cytoskeletal deregulation drives cell competition in the Drosophila eye disc. By overexpressing RhoGEF2 to induce cytoskeletal disruption and utilizing genetic knockdowns of various candidate genes, the authors examine the spatial distribution and interaction between normal and deregulated cell populations. Their findings demonstrate that cell competition and clone elimination in this context are dependent on sas-Ptp10D, scrib, and components of the Hippo signaling pathway. The study is well executed and provides a potentially impactful contribution to the field. The experimental design is solid, and the conclusions are generally well supported by the data. Only minor revisions are needed to strengthen clarity and presentation. Specific suggestions and comments regarding significance are listed below.
Major comments: There is a discrepancy between the representative images (Fig. 3A-C′) and the quantification in Fig. 3J. The statistical analysis may be limited by small sample size or suboptimal test choice (e.g., Kruskal-Wallis vs. ANOVA). Increasing the sample size and reassessing the statistical approach could strengthen this otherwise well-executed section.
Response: The data is normally distributed, so we have repeated the analysis using a one-way ANOVA (instead of the Kruskal-Wallis test – Initially we used this one because of the small sample number, but the data is normally distributed, and so a one-way ANOVA is appropriate). From examining all the images again, we can ascertain that there is indisputably less active caspase-3 staining in RhoGEF2-OE Ptp10D-KD compared to RhoGEF2-OE Dicer2. We have selected a more suitable image that better represents this snapshot of active caspase-3 staining in RhoGEF2-OE Ptp10D-KD. Also, a more representative control image is now shown, where some baseline active caspase-3 staining is present.
A minor concern relates to the interpretation and consistency of the statistical analyses used. For example, in Figure 5I, both the Kruskal-Wallis test and an unpaired t-test were used, with the authors stating that the t-test was applied specifically to compare wild-type and crb-/- clones (p = 0.0147). However, in the adjacent panel (Figure 5J), only a one-way ANOVA was used. This inconsistency may give the impression that the choice of statistical test in Figure 5I was influenced by a lack of significance with the Kruskal-Wallis test, rather than by experimental design. Unifying the statistical approach within related panels would improve clarity and minimize potential reader misinterpretation. Additionally, some of the statistical tests applied may not fully align with the underlying data distributions. Statistical methods used in parts of the manuscript may need to be reevaluated, and the rationale for their selection should be clarified in the text.
Response: We have checked the data carefully, plotted all the individual data sets in R, and the data is not normally distributed. Therefore, conducting a Kruskal-Wallis test is the best approach. This analysis shows that there is no significant difference between crb-/- and WT in our experimental setting. However, there is a slight trend towards increased crb-/- clone size. We have added a more detailed description of the statistical methods used in different situations in the Materials and Methods section.
In the section on how crb-/- affects actin distribution and accumulation within the tissue (Figure 6H′ and Supplementary Figure 5), it appears that F-actin may accumulate more prominently in cytoplasmic regions rather than at cell-cell junctions under crb-/- conditions. However, due to the current level of magnification, it is difficult to determine the precise subcellular localization. Although this question is somewhat tangential to the main focus of the manuscript and not essential for publication, it could be valuable, if the authors included a few higher-magnification images showing F-actin distribution in RhoGEF2OE Dicer2, RhoGEF2OE Ptp10D KD, and RhoGEF2OE crb-/- conditions. Including these in the supplementary figures could help clarify how actin cytoskeletal regulation is affected.
Response: We have added zoomed-in images to Figs 6G and 6H to show the effect on F-actin more clearly. It is possible that F-actin may be more prominent in the cytoplasm in crb-/- clones, however further experiments would be needed to provide more evidence for this, which are unfortunately beyond the scope of our capabilities at this time.
In Figure 6H′ the Diap1 signal in the RhoGEF2OE condition appears non-uniform, with noticeably weaker intensity on the left side of the image and stronger signal on the right. This asymmetry is not observed in the RhoGEF2OE crb-/- condition shown in Figure 6K′. It is unclear whether this pattern reflects a biological phenomenon consistently observed in RhoGEF2OE tissues or if it might result from technical factors such as uneven mounting or imaging. To prevent potential misinterpretation, we recommend clarifying this point, providing additional representative images if available, or replacing the current image with one that more clearly reflects the typical expression pattern.
Response: We assume the reviewer means Fig 6J, and we have replaced the image with a more representative one.
In Fig. 3B′, cleaved Caspase-3 appears localized to specific regions at the WT/RhoGEF2OE interface, suggesting spatial bias in Ptp10D-dependent elimination. This raises important questions about what determines regional susceptibility-are certain tissue conditions or cell states more prone to apoptosis in this context? Figure 3 raises the question of whether RhoGEF2OE-induced, actin-deregulated clones undergo dynamic changes, such as expanding or regressing, over the course of the larval stage. Such temporal variability could influence GFP⁺ clone size and the expression of apoptotic markers like cleaved Caspase-3 and Diap1. The stated use of the L3 stage, which spans ~48 hours (Tennessen & Thummel, 2011), lacks sufficient temporal resolution. Clarifying the timing of dissection and fixation relative to clone induction would improve interpretation of clone behavior and marker dynamics.
Response: While the reviewer raises an interesting question about spatial and temporal sensitivities to apoptosis upon genetic perturbations, we have conducted all of our experiments on samples obtained from the wandering L3 stage. We have added the following text to the Materials and Methods to make it clearer: “Wandering third-instar larvae (L3) were picked for all experiments, and for each experiment all larvae were of equivalent size.”.
Minor comments: GFP signal appears weaker in the wild-type group compared to experimental conditions, raising the question of whether image processing (e.g., contrast and color balance) was applied uniformly and if this difference reflects true variation in expression.
Response: Yes, images were always identically processed. We have stated in the Materials and Methods imaging section: “Laser intensity and gain was unchanged within each experimental group”.
For Figures 2, 3, and 5, including representative images for each eye phenotype category would clarify the scoring criteria. In Figure 5, the use of a "2.5" category in the main figure should be explained-does it correspond to category 3 or indicate an intermediate phenotype?
Response: Apologies for this error, and thanks to the reviewer for highlighting this. The “2.5” rating was a mistake based on a previous classification scale we used, and we have changed 2.5 to 3 in the graph. We have also included a new supplementary figure explaining our rankings (Supp Fig 10).
In Figure 5I, the y-axis range (0-150%) is broader than needed; adjusting it to 0-100% would better reflect the data and improve clarity.
Response: We have edited the Fig 5I graph accordingly.
The sentence from line 343- 348 is long and challenging to follow.
Response: We have reworded the sentence.
Missing the Figure number on Line 286.
Response: We have added the Figure number.
Reviewer #2 (Significance (Required)):
Significance: This study is well executed and rigorously addresses previously reported variations in phenotypic outcomes across laboratories. Beyond clarifying the role of Ptp10D in cell competition, the authors establish RhoGEF2 overexpression as a reliable method to induce cell competition and identify key molecular players involved in this process. This work represents a meaningful advance by introducing novel approaches and deepening understanding of known factors in clone elimination. The mosaic RhoGEF2 overexpression technique developed in this study provides a valuable tool for investigating cell-cell interactions at the tissue level, with broad applicability in basic research. This approach holds particular promise for probing.
Response: We thank the reviewer for their support of the significance and quality of our manuscript.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
In the manuscript "Actin cytoskeletal deregulation, caused by RhoGEF2 overexpression, induces cell competition dependent on Ptp10D, Crumbs, and the Hippo signaling pathway", Natasha et al. investigate how actin cytoskeletal deregulation drives cell competition in the Drosophila eye disc. By overexpressing RhoGEF2 to induce cytoskeletal disruption and utilizing genetic knockdowns of various candidate genes, the authors examine the spatial distribution and interaction between normal and deregulated cell populations. Their findings demonstrate that cell competition and clone elimination in this context are dependent on sas-Ptp10D, scrib, and components of the Hippo signaling pathway. The study is well executed and provides a potentially impactful contribution to the field. The experimental design is solid, and the conclusions are generally well supported by the data. Only minor revisions are needed to strengthen clarity and presentation. Specific suggestions and comments regarding significance are listed below.
Major comments:
There is a discrepancy between the representative images (Fig. 3A-C′) and the quantification in Fig. 3J. The statistical analysis may be limited by small sample size or suboptimal test choice (e.g., Kruskal-Wallis vs. ANOVA). Increasing the sample size and reassessing the statistical approach could strengthen this otherwise well-executed section. A minor concern relates to the interpretation and consistency of the statistical analyses used. For example, in Figure 5I, both the Kruskal-Wallis test and an unpaired t-test were used, with the authors stating that the t-test was applied specifically to compare wild-type and crb-/- clones (p = 0.0147). However, in the adjacent panel (Figure 5J), only a one-way ANOVA was used. This inconsistency may give the impression that the choice of statistical test in Figure 5I was influenced by a lack of significance with the Kruskal-Wallis test, rather than by experimental design. Unifying the statistical approach within related panels would improve clarity and minimize potential reader misinterpretation. Additionally, some of the statistical tests applied may not fully align with the underlying data distributions. Statistical methods used in parts of the manuscript may need to be reevaluated, and the rationale for their selection should be clarified in the text. In the section on how crb-/- affects actin distribution and accumulation within the tissue (Figure 6H′ and Supplementary Figure 5), it appears that F-actin may accumulate more prominently in cytoplasmic regions rather than at cell-cell junctions under crb-/- conditions. However, due to the current level of magnification, it is difficult to determine the precise subcellular localization. Although this question is somewhat tangential to the main focus of the manuscript and not essential for publication, it could be valuable, if the authors included a few higher-magnification images showing F-actin distribution in RhoGEF2OE Dicer2, RhoGEF2OE Ptp10D KD, and RhoGEF2OE crb-/- conditions. Including these in the supplementary figures could help clarify how actin cytoskeletal regulation is affected. In Figure 6H′, the Diap1 signal in the RhoGEF2OE condition appears non-uniform, with noticeably weaker intensity on the left side of the image and stronger signal on the right. This asymmetry is not observed in the RhoGEF2OE crb-/- condition shown in Figure 6K′. It is unclear whether this pattern reflects a biological phenomenon consistently observed in RhoGEF2OE tissues or if it might result from technical factors such as uneven mounting or imaging. To prevent potential misinterpretation, we recommend clarifying this point, providing additional representative images if available, or replacing the current image with one that more clearly reflects the typical expression pattern. In Fig. 3B′, cleaved Caspase-3 appears localized to specific regions at the WT/RhoGEF2OE interface, suggesting spatial bias in Ptp10D-dependent elimination. This raises important questions about what determines regional susceptibility-are certain tissue conditions or cell states more prone to apoptosis in this context? Figure 3 raises the question of whether RhoGEF2OE-induced, actin-deregulated clones undergo dynamic changes, such as expanding or regressing, over the course of the larval stage. Such temporal variability could influence GFP⁺ clone size and the expression of apoptotic markers like cleaved Caspase-3 and Diap1. The stated use of the L3 stage, which spans ~48 hours (Tennessen & Thummel, 2011), lacks sufficient temporal resolution. Clarifying the timing of dissection and fixation relative to clone induction would improve interpretation of clone behavior and marker dynamics.
Minor comments:
GFP signal appears weaker in the wild-type group compared to experimental conditions, raising the question of whether image processing (e.g., contrast and color balance) was applied uniformly and if this difference reflects true variation in expression. For Figures 2, 3, and 5, including representative images for each eye phenotype category would clarify the scoring criteria. In Figure 5, the use of a "2.5" category in the main figure should be explained-does it correspond to category 3 or indicate an intermediate phenotype? In Figure 5I, the y-axis range (0-150%) is broader than needed; adjusting it to 0-100% would better reflect the data and improve clarity. The sentence from line 343- 348 is long and challenging to follow. Missing the Figure number on Line 286.
This study is well executed and rigorously addresses previously reported variations in phenotypic outcomes across laboratories. Beyond clarifying the role of Ptp10D in cell competition, the authors establish RhoGEF2 overexpression as a reliable method to induce cell competition and identify key molecular players involved in this process. This work represents a meaningful advance by introducing novel approaches and deepening understanding of known factors in clone elimination. The mosaic RhoGEF2 overexpression technique developed in this study provides a valuable tool for investigating cell-cell interactions at the tissue level, with broad applicability in basic research. This approach holds particular promise for probing
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary: findings and key conclusions
Epithelial cell competition in larval imaginal discs involves signaling with the Sas ligand and Ptd10D receptor. In wild type cells both are typically found at the apical surface, but relocalize to the lateral cortex at the winner-loser interface. Ptd10D activation leads to reduced Ras signaling, increased pro-apoptotic Jnk signaling and consequently the elimination of loser cells. In the manuscript the authors address the role of the actin cytoskeleton in the context of the signaling controlling cell elimination in Drosophila larval eye imaginal discs. They interfere by clonal overexpression of the guanyl nucleotide exchange factor RhoGEF2 (RG2), which has previously been shown to induce dominant gain-of-function phenotypes by activation of Rho signaling. In this context the requirement of and genetic interactions with the other pathways implicated in cell elimination is tested. They find that RG2 induced cell elimination depends on PtD10D, Hippo signaling and Crumbs.
Major comments: claims and conclusions
The experimental setting, using clonal analysis in imaginal discs, is straight-forward and well-established, including quantification of clone size and comparison of phenotypes. The presented data are of high quality and thus the direct conclusions are fully supported by the data as long as they refer to the actual experimental interference. What is not supported by the data is the generalization of the conclusions, i. e. that RG2 overexpression would be equivalent to Actin cytoskeletal deregulation. This equivalence is expressed in the title "Actin cytoskeletal deregulation, caused by RhoGEF2 overexpression.." and the summary " that actin cytoskeleton deregulated cells (as induced by RhoGEF2 overexpression (RhoGEF2OE))...". In my view such an equivalence is not justified. There is no doubt that RG2 overactivation affects the actin cytoskeleton in multiple ways, such as contractility via MyoII or polymerization via Dia, among others. There is also no doubt that other pathways are also directly or indirectly affected beside the actin cytoskeleton. The authors do not present data showing the specificity of RG2 overexpression. For example, the authors could investigate the phenotype and genetic interaction with an alternative way of interference, independent of RG2, of the actin cytoskeleton to support their conclusion.<br /> There is a second assumption, which may not be justified, that the function of the cytoskeleton would be generally downstream of cell polarity, see abstract l24 "triggering cytoskeletal deregulation (which occurs downstream of cell polarity disruptions)..". There are certainly cytoskeletal activities such as cell shape changes that mediate the execution of cell elimination. However interfering with the cortical cytoskeleton also affect the distribution of cortical polarity proteins. The authors do not present data to demonstrate the specificity of RG2 overexpression concerning a function downstream of cell polarity.
The study establishes genetic interactions and dependencies concerning cell elimination following a very specific experimental interference of RG2 overexpression. It remains unclear, however, to which degree these genetic interactions contribute to controlling cell competition in situations that are physiologically relevant. The generalization of RG2 overexpression as a specific test the function of the actin cytoskeleton is an interpretation not supported by the presented data and the experimental set up.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
The authors do not wish to provide a response at this time.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
Cells need to adjust their gene expression pattern, including nutrient transporters and enzymes to process the available nutrient. How cells maintain the coordination between these processes is one of the most critical questions in biology. In this work authors elegantly combined a range of relevant experimental techniques, ranging from time-lapse microscopy, microfluidics, and mathematical modelling to address this question. Combining these methods, authors proposed a push-pull like mechanism, involving two pairs of repressors (Mth1, Std1 and Migs) in the glucose sensing network. In budding yeast there are multiple hexose transporter genes with varying affinity and transport rate. Authors postulated that on sensing glucose, cells switch between expressing high affinity glucose transporters (when extracellular glucose is low), and low affinity glucose transporters (in high extracellular glucose), and these processes are mediated by the pairs of repressors as mentioned earlier. Following the expressing patterns of fluorescently tagged hexose transporters and varying the extracellular glucose concentrations in media, authors proposed that pairs of repressors switch their activity depending on extracellular glucose level, and which is matched by the promoters of the hexose transporter genes to achieve optimality of glucose transport.
This study is elegantly designed and addressed an interesting question. The mechanism (push-pull involving two pairs of repressors) is plausible and justified by the data. Authors also presented a mathematical model and made predictions, which are also verified. We will recommend the publication of this work with minor modifications.
Major comments:
This study is well designed and experiments performed accordingly. We have only minor comments for revision.
Minor comments:
Referee cross-commenting
All other reviewers also identified this study insightful and interesting, similar to our comments. We also agree with the suggestions made by other reviewers. Suggested changes and modifications can be addressed within a month as mentioned by most of the reviewers. Excellent point raised by other reviewers on technicalities and addressing those points will improve the readability of this work even more.
General assessment:
Use of innovative microfluidics platform to trap mother cells and following the gene expression pattern by fluorescence microscopy and combining the experimental approach with mathematical model are the strengths of this work. Whereas the proposed push-pull mechanism is not generalizable to other carbons. Model is merely used to fit the data, rather than making interesting predictions. Also how does the mechanism holds when cells are switched from other nutrient sources is also not clear in this work, which are the limitations of this work.
Advance
This work involves experimental technique and mathematical model to test the hypothesis. Use of custom-built microfluidics set up and live cell imaging to track gene expression levels in varying nutrient condition. This study links single cell level gene expression pattern to model and predict system level behavior. Nutrient sensing and subsequent rearrangement of gene regulatory network is an important question to address, and the proposed push-pull mechanism in this study adds up to the existing body of literature.
Audience:
This work is interdisciplinary and researchers across multiple fields will be interested in this work, including researchers interested in microbial nutrient sensing, systems biology, topology of gene regulatory network, metabolism, and general microbiology.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
The yeast Saccharomyces cerevisiae possesses a large family of hexose transporters, the HXT genes. Some of these transporters play known roles in transport to feed metabolism while others seem to respond to glucose levels but have differing cellular functions, acting more as sensors than as drivers of carbon and energy production. The authors use single cell fluidics to monitor steady state expression of specific transporters under controlled glucose levels. The authors then used published information on the regulatory network of HXT4 gene expression to predict expression levels and confirm the role of the prior identified regulators. Thus, this work confirms prior work as to the levels of substrate leading to optimized expression of transporters and confirms the role of the identified regulatory network. The fact that the main single cell fluidics findings confirm the prior culture analyses affirms the utility of the prior work.
Major Comments:
no Minor Comments
It has been known for quite some time that glucose transport in the yeast Saccharomyces cerevisiae is dynamically regulated to optimize sugar depletion to sugar metabolism. This intricate system involves a family of hexose transporters of differing affinities for substrate, the timing and level of expression of which is regulated by both eternal hexose levels and internal ability to metabolize keeping cytoplasmic sugar levels low. Since facilitated diffusion systems can transport in both directions, the consumption of substrate assures the direction of uptake will be dominant. The authors demonstrate in this paper that differential expression of the known major regulators of HXT gene expression work in concert to adjust the expression patterns of transporters of differing affinities leading to optimization of hexose uptake. The study monitored changes in single cells and findings confirm prior work conducted in cell populations. One assumption has always been that the mother cell might "sacrifice" itself by not being able to dynamically clear the membrane of environmentally unmatched hexose transporters relying on the altered membrane composition of the bud. This work's focus on "mother cells" demonstrates that regulation still occurs if cells are allowed to reach a steady state. The timeline may be slower than bud adaptation, but these authors confirm that mother cells respond dynamically to glucose levels.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
This is a very insightful work showing how to disentangle one of the most complex transcriptional networks in yeast (S. cerevisiae) by combining single-cell dynamics, dynamical-systems modeling, Bayesian-style inference, and genetic perturbations. The authors tackle a problem that has eluded quantitative resolution for over two decades-how yeast regulates its seven primary glucose importer genes (HXT1-HXT7) in response to both steady and temporally changing extracellular [glucose]. Their integrated experimental-theoretical approach delivers the most satisfying mechanistic and quantitative explanation to date, and I enthusiastically recommend this manuscript for publication.
Yeast relies on seven passive hexose transporters (Hxt1-Hxt7) to import glucose, its preferred sugar; deleting all seven abolishes growth on glucose. The underlying regulatory network is exceptionally intricate, reflecting yeast's evolutionary priority for glucose. Two membrane sensors-Snf3 (high affinity) and Rgt2 (low affinity)-detect extracellular glucose and thereby inactivate two co-repressors, Mth1 and Std1, which modulate the DNA-binding factor Rgt1. Concurrently, intracellular glucose activates the SNF1 kinase, phosphorylating and exporting the repressor Mig1, while Mth1/Std1 also govern the transcription and stability of Mig2, another DNA-binding repressor. Together, Rgt1, Mig1, and Mig2 integrate these inputs to control HXT promoter activity (Fig. 2A). Importantly, Mth1 and Std1 do not directly bind to DNA and this complication - the protein-protein interaction that one cannot get from DNA sequence - is just one source of difficulty that the authors overcame.
To map the network's behavior, the authors used microfluidic "cages" housing single cells expressing GFP-tagged HXTs, monitoring fluorescence under three constant glucose levels-low (0.01%), medium (0.1%), and high (1%) (Fig. 1B-C). The authors confirm that steady-state Hxt abundances rank by transporter affinity. But the more important and surprising discovery is that when the cells were subjected to gradual glucose up-shifts and down-shifts, they discovered that some transporters transiently spike only when [glucose] rises and others only when [glucose] falls (Fig. 1C and Fig. S1F). This discovery establishes that the HXT network not only "senses" the absolute external [glucose] concentration but also the direction of the temporal change in external [glucose].
To understand how the regulatory network yields such intricate temporal changes in HXT expression, the authors first focused on the medium-affinity transporter, Hxt4. Targeted knockouts of Mig1/Mig2 versus Mth1/Std1 confirmed that Hxt4 dynamics arise from differential repressor kinetics. To formalize these findings, the authors built an ODE model grounded in literature-based constraints (pg. 13 of the Supplement) with explicit separation of repressor timescales. They rigorously fit the model to wild-type and knockout time series-exploring parameter sensitivity in depth (Fig. S5).
The authors discovered that their model and experiments converged on a push-pull mechanism: fast-acting Mig1/Mig2 dominate during glucose up-shifts, while slower Mth1/Std1 govern down-shifts, determining whether each HXT gene is repressed or de-repressed (i.e., "who gets there first"). Extending this analysis across all seven HXTs via approximate Bayesian computation revealed the most likely repressor-promoter interactions for each transporter, reducing a vast parameter space to unique or small sets of plausible regulatory schemes. The authors thus revealed what could be happening and which regulations are improbable - a more nuanced and comprehensive view than giving just one outcome for each HXT.
Overall, this work represents a role model - textbook-worthy - for quantitative systems biology. Beyond the rigor and novelty of its findings, the authors explain complex mathematical concepts with clarity, and the narrative flows logically from experiment to model to inference. This study provides a definitive mechanistic resolution of the HXT network and establishes a broadly applicable framework for dissecting dynamic and complex gene circuits.
Major points:
I don't recommend any new experiments or modeling; the major claims are already well supported by the data and models. Below are comments and questions intended to improve clarity and facilitate the reader's understanding. Please feel free to disregard any that you find not sensible or beyond the scope of the current work.
Minor points:
Hyun Youk.
Referee cross-commenting
I agree with the other reviewers' comments. The other reviewers noticed important points I have missed. But like them, I'm still supportive of the work being published with < 1 month spent on revision. I still don't recommend any further experiments or modeling.
This is a very insightful work showing how to disentangle one of the most complex transcriptional networks in yeast (S. cerevisiae) by combining single-cell dynamics, dynamical-systems modeling, Bayesian-style inference, and genetic perturbations. The authors tackle a problem that has eluded quantitative resolution for over two decades-how yeast regulates its seven primary glucose importer genes (HXT1-HXT7) in response to both steady and temporally changing extracellular [glucose]. Their integrated experimental-theoretical approach delivers the most satisfying mechanistic and quantitative explanation to date, and I enthusiastically recommend this manuscript for publication via Review Commons.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Manuscript number: RC-2025-03083 Corresponding author(s): David Fay General Statements [optional] This section is optional. Insert here any general statements you wish to make about the goal of the study or about the reviews.
We greatly appreciate the input of the four reviewers, all of whom carried out a careful reading of our manuscript, provided useful suggestions for improvements, and were enthusiastic about the study including its thoroughness and utility to the field. Because the reviewers required no additional experiments, we were able to address their comments in writing.
However, in response to a comment from reviewer #4 we decided to add an additional new biological finding to our study given that our functional validation of proximity labeling targets was not extensive. Namely, we now show that a missense mutation affecting BCC-1, one of the top NEKL-MLT interactors identified by our proximity labeling screen, is a causative mutation (together with catp-1) in a strain isolated through a forward genetic screen for suppressors of nekl molting defects (new Fig 9C). This finding, combined with our genetic enhancer tests, further strengthens the functional relevance of proteins identified though our proximity labeling approach and highlights the synergy of proteomics combined with classical genetics.
Positive statements from reviewers include: Reviewer #1: Overall, this is an outstanding study that will be of great interest to those interested in using proximity labeling to identify interactors of their favorite protein. The experiments are well executed and the data presented in a mostly clear manner.
Reviewer #2: The key conclusions are convincing, and the work is rigorous. The work provides a clear roadmap to reproducing the data. The experiments are adequately replicated, and statistical analysis is adequate... In many papers, TurboID seems very trivial but this paper clearly highlights the limitations and will be an invaluable resource for labs that want to get proximity labeling established in their labs.
Reviewer #3: Overall, the claims are solid and conclusions supported. The data and methods are substantial to enable reproducibility in other labs. The experiments have been repeated multiple times with particular attention to statistical analysis. ...This manuscript represents a methodological advance that will likely become an oft-cited reference for members of the C. elegans community and a springboard for other basic biomedical scientists wanting to adapt rigorous proximity labeling techniques to their system.
Reviewer #4: Fay et al. present a solid, clear and comprehensive BioID-based proteomics study that takes into account and discusses decisive aspects for the (re)production and analysis of high-quality TurboID-based mass spectrometry data. Claims and conclusions are generally well and sufficiently supported by the presented data and illustrated with figures (throughout the text as well as with plenty of supplementary data)... Basic consideration and thoughts for the experimental design and MS data analysis are given in detail and can serve as another guideline for future studies.
Based on these reviews and comments, we believe that our manuscript is suitable for publication in a high-impact journal. 1. Point-by-point description of the revisions This section is mandatory. Please insert a point-by-point reply describing the revisions that were already carried out and included in the transferred manuscript.
*Reviewer #1 (Evidence, reproducibility and clarity (Required)): *
*Proximity labeling has become a powerful tool for defining protein interaction networks and has been utilized in a growing number of multicellular model systems. However, while such an approach can efficiently generate a list of potential interactors, knowledge of the most appropriate controls and standardized metrics to judge the quality of the data are lacking. The study by Fay systematically investigates these questions using the C. elegans NIMA kinase family members NEKL-2 and NEKL-2 and their known binding partners MLT-2, MLT-3 and MLT-4. The authors perform eight TurboID experiments each with multiple NEKL and MLT proteins and explore general metrics for assessing experimental outcomes as well as how each of the individual metrics correlates with one another. They also compare technical and biological replicates, explore strategies for identifying false positives and investigate a number of variations in the experimental approach, such as the use of N- versus C-terminal tags, depletion of endogenous biotinylated proteins, combining auxin-inducible degradation, and the use of gene ontology analysis to identify physiological interactors. Finally, the authors validate their findings by demonstrating that a number of the candidate identified functionally interact with NEKL-2 or components of the WASH complex. *
Overall this is an outstanding study that will be of great interest to those interested in using proximity labeling to identify interactors of their favorite protein. The experiments are well executed and the data presented in a mostly clear manner. I really like this study (particularly because I plan to do a proximity labeling study of my own), but I did come away less than impressed with some of the analysis. This is a data-dense manuscript, and it appears to me that the authors tried to cover so much ground that in some cases very little insight was provided. For instance, the authors promote the use of data independent acquisition (DIA) as compared to the more commonly used data dependent acquisition (DDA). However the authors do not provide any analysis to indicate one approach is better than the other. Likewise the combined use of auxin-induced degradation and proximity labeling is explored but there is very little to take away from these experiments. Despite these issues, I am very enthusiastic about the study as a whole. Below I list major and minor concerns.
Major concerns * 1. My biggest issue with the manuscript is that a lot is made of the use of data independent acquisition (DIA) as compared to the more commonly used data dependent acquisition (DDA). The authors perform experiments using DIA and DDA approaches but do not directly compare the outcomes. As a result there is really no way to know if one approach is better than the other. I would suggest the authors either perform the necessary analysis to compare the two approaches or tone down their promotion of DIA.* We agree and have scaled back any statements comparing DDA to DIA as our manuscript did not address this directly. We also now point out this caveat in our closing thoughts section, while referencing other studies that compared the two (lines 926-929). Our main point was to convey that DIA worked well for our proximity labeling studies but has seen little use by the model organism field. Surprising (to us), DIA was also considerably less expensive than DDA options.
2. Line 75, The authors promote the use of data-independent acquisition (DIA) without defining what this approach is and how it differs from the more conventional data-dependent acquisition. As a non-mass spectroscopist, I found myself with lots of question concerning DIA, what it is and how it differs from DDA. I think it would really be helpful to expand the description of DIA and its comparison with DDA in the introduction. As non-mass-spectroscopists ourselves, we understand the reviewer's point. Because the paper is quite long, we were trying to avoid non-essential information. We have now added some information to explain some of the key differences between DDA and DIA. We have also included references for readers who may want to learn more. (lines 77-80)
Minor concerns: * Line 92 typo. I believe the authors meant to say NEKL-2-MLT-2-MLT-4. * Corrected. (line 95)
Line169. Is exogenous the correct word to use here? It suggests that you are talking about non-worm proteins, but I know you are not. Corrected. Changed to "Moreover, the detection of biotinylated proteins may be difficult if the bait-TurboID fusion is expressed at low levels..." (line 181).
Line 177 typo (D) should be (C). Corrected. (line 1122)
Figure 1C: Lucky Charms may sue you for infringement of their trademarked marshmallow treats. Thank you for picking up on this. The authors accept full responsibility for any resulting lawsuits.
Figure 1D. The NEKL-2::TurboID band is indicated with a green triangle in the figure but the figure legend states that green triangles indicate mNG::TurboID control. I know this triangle is a shade off the triangle that indicates mNG::TurboID but it's really hard to see the difference. All of the differently colored triangles in panel F are unnecessary. I would either just pick one color for all non-control bait proteins or better yet, only use a triangle to point to bands that are not obvious. For instance I don't need the triangles that point to NEKL-2 -3 and -4 fusion proteins. These are just distracting. We understand the reviewer's point. We colored the triangles to match the colors used for the proteins in the figures. We have now added "bright green triangles with white outlines" (Fig 1 legend) to indicate the Pdpy-7::mNG::TurboID control" and changed triangles in the corresponding figures. Although we would be fine with removing or changing the triangles, we think that they may aid somewhat with clarity.
Line: 316: Conceivably, another factor that could contribute to the counterintuitive upregulation of some proteins in the N2 samples is related to the fusion proteins that are being expressed in the TurboID lines. A partially functional bait protein (one with a level of activity similar to nekl-2(fd81) that may not result in an obvious phenotype) could directly or indirectly affect gene expression leading to lower levels of a subset of proteins in the TurboID samples. The same could be said for fusion proteins with a gain-of-function effect. This is an interesting idea, and we tested this possibility by looking for consistent overlap between N2-up proteins between biological replates of individual bait proteins. We now include a representative Venn diagram in S3C Fig to highlight this comparison. In summary, although we cannot rule out this possibility, our analysis did not support the widespread occurrence of this effect in our study. We also made certain that our statement regarding N2 up proteins was not too definitive. (lines 285-288)
*Fig 3 B-E. I am a little confused how the data in these graphs is normalized. For instance, I would have expected that for NEKL-3 in panel B, that the normalized (log2) intensity value in N2 be set at 0 as it is for NEKL-2. Maybe I just don't have enough information on how these plots were generated. * The difference is that in the N2 sample, NEKL-3 was detected but NEKL-2 was not. The numbers themselves are assigned by the Spectronaut software used to quantify the DIA results but are not meaningful beyond indicating relative amounts (intensity values) of a given protein within an individual biological experiment. We've added some lines to the figure legend to make this clearer. (lines 1165-1169)
*Figure 6C legend is not correct. * Corrected. (line 1214)
Line 575: Figure reference should be Fig. S5G. The authors should check to make sure all references to supplemental figures include correct panel information. Corrected. (line 464) In addition, we have now gone through the manuscript and added panel numbers references where applicable. Note that the addition of a new supplemental file has shifted the numbering.
Line 576. The authors reference a study by Artan and colleagues and report a weak correlation between their study and that of Artan. They reference figure S4 but it should be Fig S5H. Apologies and many thanks to the reviewer for catching these errors. (line 464)
Line 652. The authors note that numerous proteins were present at substantially reduced levels in the mNG::TurboID samples and suggest that sticky proteins may have been outcompeted or otherwise excluded from beads incubated with the mNG::TurboID lysates. Why would sticky proteins only be a problem in these samples? The reasoning is not clear to me. The idea was that in the sample with very high levels of biotinylated proteins (mNG::TurboID), the surface of the beads might become saturated with high-affinity biotinylated proteins. This could prevent or out complete the binding of random proteins that are not biotinylated but nevertheless have some affinity to the beads ("sticky" proteins). We have reworded this section to make this clearer. (lines 546-550)
Line 745: The term "bait overlaps" is a bit vague. Ultimately, I figured out what it meant but it was not immediately obvious. We have changed this to "overlap between baits" and made this section clearer. (line 624-628)
*S7B Fig. Why is actin missing from the eluate? * In S7B we refer to the purified eluate as the "eluate", which may have caused some confusion. In other sections of the manuscript, we refer to the bead-bound proteins as the "purified eluate" (Figs 1 and 5). For the purified eluate a portion of the streptavidin beads are boiled in sample buffer to elute the bound proteins before running a western. Actin would not be expected in these samples because it's (presumably) not biotinylated in our samples and doesn't detectably bind the beads. This result was seen in all relevant westerns in S1 Data. For consistency, however, we've gone through all our files to make sure we consistently use the term "purified eluate" versus "eluate", which is less specific.
L*ine 873: The authors state the extent of overlap in GO terms between the various experiments and provide percentages. I tried to extract this information from Figure 8C and came up with different values. For instance, in the case of Molecular Function, they state that they observed a 54% overlap between NEKL-2 and NEKL-3 but in the Venn diagram in Figure 8C I see that the NEKL-2 and NEKL-3 experiments had 71 (25+46) GO terms in common. Out of 98 GO terms for NEKL-2 or 104 for NEKL-3 the percentage I got is closer to 72. Am I analyzing this correctly? * Thanks for checking this. We believe our method for calculating the percent overlap is correct. In the case of NEKL-2/NEKL-3 overlap for Molecular Function, there are 131 total unique terms, of which 71 overlap, giving a 54% overlap. In the case of NEKL-2/NEKL-3 overlap for Biological Process, however, we made an error in arithmetic (415 unique, 239 overlap), such that the correct percentage is 58%, which we have corrected in the text.
*Reviewer #1 (Significance (Required)): *
*Overall this is an outstanding study that will be of great interest to those interested in using proximity labeling to identify interactors of their favorite protein. The experiments are well executed and the data presented in a mostly clear manner. I really like this study (particularly because I plan to do a proximity labeling study of my own), but I did come away less than impressed with some of the analysis. This is a data-dense manuscript, and it appears to me that the authors tried to cover so much ground that in some cases very little insight was provided. For instance, the authors promote the use of data independent acquisition (DIA) as compared to the more commonly used data dependent acquisition (DDA). However the authors do not provide any analysis to indicate one approach is better than the other. Likewise the combined use of auxin-induced degradation and proximity labeling is explored but there is very little to take away from these experiments. Despite these issues, I am very enthusiastic about the study as a whole. *
*Reviewer #2 (Evidence, reproducibility and clarity (Required)): *
*This study expanded the use of data-independent acquisition-mass spectrometry (DIA-MS) in TurboID proximity-labeling proteomics to identify novel interactors of NEKL-2, NEKL-3, MLT-2, MLT-3, and MLT-4 complexes in C. elegans. The authors described several useful metrics to evaluate the quality of TurboID experiments, such as using the percentage of upregulated genes, the percentage of proteins present only in bait-TurboID experiments as compared to N2 controls, and the percentage of endogenously biotinylated carboxylases as internal controls. Further, the authors introduced methodological variability across 23 TurboID experiments and evaluated any improvement to the resulting data, such as N-terminally tagging bait proteins with TurboID, depleting endogenous carboxylases, and auxin-inducible degradation of known complex members. Finally, this study identified the kinase folding chaperone CDC-37 and the WASH complex component DDL-2 as novel interactors with the NEKL-MLT complexes through an RNAi-based enhancer approach following their identification by TurboID. *
Major comments: * The key conclusions are convincing, and the work is rigorous. The work provides a clear roadmap to reproducing the data. The experiments are adequately replicated, and statistical analysis is adequate. We only have minor comments.*
Minor comments: * •In the western blot in Fig 1 why does the mNG::Turbo have two bands? * Thank you for point this out. To our knowledge this is a breakdown product that was especially prevalent in replicate 3 (also see S1 Data), which we chose to shown because all the NEKL-MLTs were clearly visible in this western. The expected size of the mNeonGreen::TurboID (including linker and tags) is ~68 kDa and our blots are roughly consistent those of Artan et al., (2001). This lower band was not evident in Exp 8. We have now included a statement in the figure legend to indicate that the upper band is the full-length protein whereas the lower band is likely to be a breakdown product (lines 1141-1142).
•Fig 2B is difficult to parse as a reader. Columns labeled "Upreg," "Downreg," "TurboID only," "N2 only," "Filter-1," "Filter-2," and "Epi %" could be moved to Supplemental. Fold change vs N2 could be represented as a bar chart, allowing for trends between fold change and the metrics Upreg %, Turbo %, and Carboxylase % to be seen more clearly. Further, rows headed "Carboxylase depletion," "DDA," and "Auxin treated" could be presented as separate panels to better match the distinct points made in the text. After serious consideration we have made several changes including the addition of S2 Fig, which may provide readers with a better visual representation of the bait and prey fold changes observed in all our experiments. However, we feel that the detailed data embedded in Fig 2 is the most concise and accurate means by which to convey our full results and is key to our methodological conclusions. As such we did not want to relegate this information to a supplemental table. We note that this figure was not found to be problematic by other reviewers, although we do understand the points made by this reviewer.
•Line 179: in vivo should be italicized Because journals differ in their stylistic practices, we are currently waiting before doing our final formatting. We did keep our use of Latin phrases consistently non-italicized in the draft.
•Lines 215-217: The comparison between Western blot expression levels and prior fluorescent reporter levels is unclear. Could be reformatted to make it clearer that relative expression of the different NEKL-MLTs in this study is consistent with prior data. We reformatted this sentence to improve clarity. (lines 205-207)
*•Lines 267-268: The final line of the passage is unclear and can be removed. * This sentence has been removed.
•Lines 311-313: This study is able to use the recovery of bait and known interactor proteins as internal controls to determine the quality of each experiment, but this may not always be the case for other users' experiments. The authors should comment on how Upreg %, a value influenced by many factors, can actually be used as a quality check when a bait protein has no known interactors. We have added language to highlight this point. (lines 344-348)
*•Line 702: There is a [new REF] that should be removed * As described above, we have now included this finding on bcc-1 as part of this manuscript (Fig 9C).
•The approach used mixed stage animals, but some genes oscillate or are transiently expressed. Please discuss cost-benefit of mixed stage vs syncing. This is an important point. We have added a discussion on the benefits and drawbacks of using mixed stages to the discussion. (lines 901-911)
*•Authors were working on hypodermally expressed proteins. It would be valuable to discuss what tissues are amenable to TurboID. Ie are the cases where there are few cells (anchor cell, glial sockets, etc) that it will be extremely challenging to perform this technique * We agree that certain tissues/proteins will not be amenable to proximity labeling. We believe that we have addressed this point together with the above comment throughout the manuscript and now on lines 936-940.
•Authors mention approaches such as nanobodies, split Turbo. Based on their experiences it would be valuable to add Discussion on strengths and weaknesses of these approaches to guide folks considering TurboID and DIA-MS experiments in C. elegans Because we have not tested these methods, we feel that we cannot provide a great deal of insight into these alternate approaches. We mention and reference these methods in the introduction so that readers are aware of them.
*Reviewer #2 (Significance (Required)): *
•Advance in technique: This study expands the use cases of data-independent acquisition MS method (DIA-MS) in C. elegans, which fragments all ions independent of the initial MS1 data. The benefits of this approach include better reproducibility across technical replicates and better recovery of low abundance peptides, which are critical for advancing our ability to capture weak and transient interactions.
•The use of DIA-MS in this study has improved our understanding of the partners of these NEKL-MLTs in membrane trafficking, molting, and cell adhesion within the epidermis.
•In many papers, TurboID seems very trivial but this paper clearly highlights the limitations and will be an invaluable resource for labs that want to get proximity labeling established in their labs.
*Reviewer #3 (Evidence, reproducibility and clarity (Required)): *
*Summary: *
Fay and colleagues perform a series of proximity labeling experiments in C. elegans followed by thorough and rational analysis of the resulting biotinylated proteins identified by LC-MS/MS. The overall goals of the study are to evaluate different techniques and provide practical guidance on how to achieve success. The major takeaways are that integration of data-independent acquisition (DIA) along with comparison of endogenously tagged TurboID alleles to soluble TurboID expressed in the same tissue results in improved detection of bona-fide interactors and reduced numbers of false-positives.
*Major comments: *
Overall the claims are solid and conclusions supported. The data and methods are substantial to enable reproducibility in other labs. The experiments have been repeated multiple times with particular attention to statistical analysis. I have no major concerns with the manuscript and focus primarily on improving the accessibility of this important contribution to the scientific community. As such, I suggest that the authors:
1) Provide more explanation of and rationale for using DIA. This is not yet a standard technique and most basic biomedical scientists will be unaware of the jargon. As I expect many labs in the C. elegans community and beyond will be interested in the guidance provided in this manuscript, the introduction offers a great opportunity to bring the reader up to speed, as opposed to sending them to the complicated proteomics analysis literature. We have added some additional context (lines 77-80) as well as new references. We note that getting into the technical differences between DIA and DDA, beyond what we briefly mention, would take a substantial amount of space, may not be of interest to many readers, and can be found through standard internet and (sigh) AI-based searches.
*2) Provide a better overview of the various protocols tested (Experiments 1-8). Maybe at the beginning of the results, and maybe with an accompanying schematic. As currently written, it is difficult to figure out details regarding how the experiments vary and why. * We have now added a short paragraph to better inform the reader at the front end regarding the major experiments. (lines 139-146).
3) As to be expected, expression of TurboID tags at endogenous levels via low abundance proteins in a complex multicellular system results in somewhat weak signals that flirt with the limit of detection. Perhaps by combining tagged alleles within the same complex (NEKL-3/MLT-3 or NEKL-2/MLT-2/MLT-4) the signals could be boosted? Tandem tags, either on one end or multiple ends of proteins might help as well. As the authors point out, a benefit of tagging the two NEKL-MLT complexes is that there are strong loss-of-function phenotypes (lethal molting defects) to help evaluate whether a tagging strategy results in a non-functional complex. THESE EXPERIMENTS ARE OPTIONAL and might simply be discussed at the authors discretion. These are interesting ideas that we have now incorporated into our discussion. (lines 936-940)
*Minor Comments: *
*1) Figure 3A is cropped on the right. * Thank you for catching this. Corrected.
*2) Better define [new REF] on line 702. * We have added new results (Fig 9C), obviating the need for this reference.
***Referee cross-comments** *
Overall, I am in agreement with, and supportive of, the other reviewers' comments.
*Reviewer #3 (Significance (Required)): *
*Significance: *
Proximity labeling is often proposed as a technique to determine interaction networks of proteins in vivo, but in practice it remains challenging for most labs to execute a successful experiment, especially within the context of multicellular model organisms. Fay and colleagues provide a much needed roadmap for how to best approach proximity labeling experiments in C. elegans that will likely apply to other model systems.
They establish a rigorous approach by choosing to endogenously tag components of two essential NEKL-MLT complexes required for C. elegans molting. These complexes are relatively low abundance as they are only expressed in a single cell type, the hyp7 epidermal syncytium. In addition, as inactivation of any member of the complexes results in molting defects, they have a powerful selection for functional tags. Thus, they have set a high bar for themselves in order to discern whether a given variation on the experimental approach results in improved detection of interactors and fewer false positives.
*Potential areas for improvement include lowering the expression level of the skin-specific soluble TurboID used to determine non-specific biotinylation events. This control results in much higher levels of biotinylation compared to the TurboID-tagged NEKL-MLT alleles and likely affects their analysis, which they openly admit. In addition, to reduce the high level of background biotinylation signals generated by endogenous carboxylases, they adopt a depletion strategy pioneered by other researchers but this does not offer major improvements in detection of specific signals. The source of these conflicting results remains to be determined. It is also curious that auxin-inducible degradation of components of the NEKL-MLT complexes did not robustly alter the resulting biotinylating capacity of other members. This approach should be evaluated in subsequent studies. Finally, as mentioned in Major Comment #3 (above), it would be interesting to see if combining TurboID tags within the same complex might improve signal-to-background ratios. *
This manuscript represents a methodological advance that will likely become an oft-cited reference for members of the C. elegans community and a springboard for other basic biomedical scientists wanting to adapt rigorous proximity labeling techniques to their system. I am a cell biologist that uses a variety of genetic, molecular and biochemical approaches, mostly centered around C. elegans. I have used LC/MS-MS in our studies but have relatively little expertise in evaluating all aspects of proteomic pipelines.
*Reviewer #4 (Evidence, reproducibility and clarity (Required)): *
*Fay et al. describe an extensive proximity labeling BioID study in C. elegans with TurboID and DIA-LCMS analysis. They chose the NEKL-2/3 kinases and their known interactors MLT-2/3/4 as TurboID-fused bait proteins (C- and partially N-terminal fusions encoded from CRISPR-mediated genome edited genes). With eight biological replicates (and three to four technical replicates each) and with the unmodified wildtype or mNeonGreen-TurboID expressing worms as controls, a comprehensive dataset was generated. Although starting from quite different abundances of the bait-fusions within the cell lysates all bait proteins and known complex-binding partners were convincingly enriched with capturing streptavidin beads after only one hour of incubation with the lysate. This confirms the general applicability of TurboID-BioID approach in C. elegans. The BioID method typically gives rise to large proteomics datasets (up to more than thousand proteins identified after biotin capture) with several tens to hundreds enriched proteins (against negative control strains) as potential proteins that localize proximal to the bait-TurboID protein. However, substantial variations of candidates between biological replicates are frequently observed in BioID experiments. The authors scrutinized their dataset towards indicative metrics, filters and cutoffs in order to separate high-confidence from low-confidence candidates. With the workflow applied the authors melt down the number of candidates to 15 proteins that were grouped in four functional groups reasonably associated to NEKL-MLT function. *
Successful BioID experiments depend on reliable enrichment quantification with mass spectrometry using control cell lines that require a carefully bait-tailored design. Those must adequately express TurboID controls matching the abundance of the bait-TurboID fusion protein and its biotinylation activity. After affinity capture, sample preparation and LCMS data acquisition there is no silver bullet towards the identification true bait neighbors. Fay et al. elaborately describe their considerations and workflow towards high-confidence candidates. The workflow considered (i) data analysis with Volcano plots to account for statistical reproducibility of biological replicates against negative controls, (ii) fraction of proteins only detected in the positive or negative controls thus evading the fold-enrichment quantification approach, (iii) evaluation of variations in carboxylase enrichment as a measure for variations in the general biotin capture quality between experiments, (iv) an assessment of technical reproducibility with scatter plots and Venn diagrams, (v) exclusion of potentially false positives, e.g. promiscuously biotinylated non-proximal proteins, through comparisons with control worms expressing a non-localized mNeonGreen-TurboID fusion protein, (vi) batch effects, (vii) the impact of endogenous biotinylated carboxylases through depletion, (viii) gene ontology analysis of enriched proteins, (ix) weighing data according to the quality of individual experiments according to the afore mentioned metrics, and finally (x) genetic interaction studies to functionally associate high-confidence candidates with the bait.
*Major comments: *
Fay et al. present a solid, clear and comprehensive BioID-based proteomics study that takes into account and discusses decisive aspects for the (re)production and analysis of high-quality TurboID-based mass spectrometry data. Claims and conclusions are generally well and sufficiently supported by the presented data and illustrated with figures (throughout the text as well as with plenty of supplementary data). However, although the authors claim to seek for substrates of the kinase complex they drew no further attention to the phosphorylation status of the captured proteins. Haven't the MS data been analyzed in this respect? Information regarding this issue would enhance the manuscript. Data generation and method description appear reproducible for readers. Also, the statistical analyses appear adequate. The authors should also consider to deposit their MS raw and analysis data in a public repository (e.g. PRIDE) for future reviewing processes and as reference data for readers and followers. Our raw MS data have been deposited by the Arkansas Proteomics Facility. I have followed up to ensure that they are publicly available.
*Minor comments: *
The authors should combine supplementary data files to reduce the number of single files readers have to deal with. We have combined these files as suggested.
The authors should avoid the term "upregulation" or "increased biotinylation" when capture enrichment is meant. We agree with reviewer's point. We now use the terms enriched versus reduced or up versus down, depending on the context, and clearly define these terms. These changes have been incorporated throughout the manuscript.
*Reviewer #4 (Significance (Required)): *
The manuscript presents a robust BioID proteomics screening for co-localizing proteins of NEKL-2/3 kinases and their known interactors MLT-2/3/4. The ongoing validation of their functional interactions and whether the protein candidates reflect phosphorylation substrates or else remains elusive and is announced for upcoming manuscripts. The knowledge gain in terms of molecular mechanisms with NEKL-2/3 MLT-2/3/4 involvement in C. elegans is therefore limited to a table of - promising - interacting candidates that have to be studied further. Information about the phosphorylation status of the captured proteins from the MS data are not given. However, knowing the protein candidates will be of interest for groups working with these complexes (or the identified potentially interacting proteins) either in C. elegans or any other organism. Also, in-depth proteomics screenings with novel approaches such as BioID have to be established for individual organisms. For C. elegans there is only one prior BioID publication (Holzer et al. 2022). Many of the aspects discussed here have also been addressed earlier for BioIDs in other organisms and are not principally new. However, the presented study can be of conceptual interest for labs delving into or entangled with the BioID method in C. elegans or other organisms. The study addresses especially proteomics groups working on protein-protein interactions using proximity labeling/MS approaches. Basic consideration and thoughts for the experimental design and MS data analysis are given in detail and can serve as another guideline for future studies.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Fay et al. describe an extensive proximity labeling BioID study in C. elegans with TurboID and DIA-LCMS analysis. They chose the NEKL-2/3 kinases and their known interactors MLT-2/3/4 as TurboID-fused bait proteins (C- and partially N-terminal fusions encoded from CRISPR-mediated genome edited genes). With eight biological replicates (and three to four technical replicates each) and with the unmodified wildtype or mNeonGreen-TurboID expressing worms as controls, a comprehensive dataset was generated. Although starting from quite different abundances of the bait-fusions within the cell lysates all bait proteins and known complex-binding partners were convincingly enriched with capturing streptavidin beads after only one hour of incubation with the lysate. This confirms the general applicability of TurboID-BioID approach in C. elegans. The BioID method typically gives rise to large proteomics datasets (up to more than thousand proteins identified after biotin capture) with several tens to hundreds enriched proteins (against negative control strains) as potential proteins that localize proximal to the bait-TurboID protein. However, substantial variations of candidates between biological replicates are frequently observed in BioID experiments. The authors scrutinized their dataset towards indicative metrics, filters and cutoffs in order to separate high-confidence from low-confidence candidates. With the workflow applied the authors melt down the number of candidates to 15 proteins that were grouped in four functional groups reasonably associated to NEKL-MLT function.
Successful BioID experiments depend on reliable enrichment quantification with mass spectrometry using control cell lines that require a carefully bait-tailored design. Those must adequately express TurboID controls matching the abundance of the bait-TurboID fusion protein and its biotinylation activity. After affinity capture, sample preparation and LCMS data acquisition there is no silver bullet towards the identification true bait neighbors. Fay et al. elaborately describe their considerations and workflow towards high-confidence candidates. The workflow considered (i) data analysis with Volcano plots to account for statistical reproducibility of biological replicates against negative controls, (ii) fraction of proteins only detected in the positive or negative controls thus evading the fold-enrichment quantification approach, (iii) evaluation of variations in carboxylase enrichment as a measure for variations in the general biotin capture quality between experiments, (iv) an assessment of technical reproducibility with scatter plots and Venn diagrams, (v) exclusion of potentially false positives, e.g. promiscuously biotinylated non-proximal proteins, through comparisons with control worms expressing a non-localized mNeonGreen-TurboID fusion protein, (vi) batch effects, (vii) the impact of endogenous biotinylated carboxylases through depletion, (viii) gene ontology analysis of enriched proteins, (ix) weighing data according to the quality of individual experiments according to the afore mentioned metrics, and finally (x) genetic interaction studies to functionally associate high-confidence candidates with the bait.
Major comments:
Fay et al. present a solid, clear and comprehensive BioID-based proteomics study that takes into account and discusses decisive aspects for the (re)production and analysis of high-quality TurboID-based mass spectrometry data. Claims and conclusions are generally well and sufficiently supported by the presented data and illustrated with figures (throughout the text as well as with plenty of supplementary data). However, although the authors claim to seek for substrates of the kinase complex they drew no further attention to the phosphorylation status of the captured proteins. Haven't the MS data been analyzed in this respect? Information regarding this issue would enhance the manuscript. Data generation and method description appear reproducible for readers. Also, the statistical analyses appear adequate. The authors should also consider to deposit their MS raw and analysis data in a public repository (e.g. PRIDE) for future reviewing processes and as reference data for readers and followers.
Minor comments:
The authors should combine supplementary data files to reduce the number of single files readers have to deal with. The authors should avoid the term "upregulation" or "increased biotinylation" when capture enrichment is meant.
The manuscript presents a robust BioID proteomics screening for co-localizing proteins of NEKL-2/3 kinases and their known interactors MLT-2/3/4. The ongoing validation of their functional interactions and whether the protein candidates reflect phosphorylation substrates or else remains elusive and is announced for upcoming manuscripts. The knowledge gain in terms of molecular mechanisms with NEKL-2/3 MLT-2/3/4 involvement in C. elegans is therefore limited to a table of - promising - interacting candidates that have to be studied further. Information about the phosphorylation status of the captured proteins from the MS data are not given. However, knowing the protein candidates will be of interest for groups working with these complexes (or the identified potentially interacting proteins) either in C. elegans or any other organism. Also, in-depth proteomics screenings with novel approaches such as BioID have to be established for individual organisms. For C. elegans there is only one prior BioID publication (Holzer et al. 2022). Many of the aspects discussed here have also been addressed earlier for BioIDs in other organisms and are not principally new. However, the presented study can be of conceptual interest for labs delving into or entangled with the BioID method in C. elegans or other organisms. The study addresses especially proteomics groups working on protein-protein interactions using proximity labeling/MS approaches. Basic consideration and thoughts for the experimental design and MS data analysis are given in detail and can serve as another guideline for future studies.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
Fay and colleagues perform a series of proximity labeling experiments in C. elegans followed by thorough and rational analysis of the resulting biotinylated proteins identified by LC-MS/MS. The overall goals of the study are to evaluate different techniques and provide practical guidance on how to achieve success. The major takeaways are that integration of data-independent acquisition (DIA) along with comparison of endogenously tagged TurboID alleles to soluble TurboID expressed in the same tissue results in improved detection of bona-fide interactors and reduced numbers of false-positives.
Major comments:
Overall the claims are solid and conclusions supported. The data and methods are substantial to enable reproducibility in other labs. The experiments have been repeated multiple times with particular attention to statistical analysis. I have no major concerns with the manuscript and focus primarily on improving the accessibility of this important contribution to the scientific community. As such, I suggest that the authors:
Minor Comments:
Referee cross-comments
Overall, I am in agreement with, and supportive of, the other reviewers' comments.
Proximity labeling is often proposed as a technique to determine interaction networks of proteins in vivo, but in practice it remains challenging for most labs to execute a successful experiment, especially within the context of multicellular model organisms. Fay and colleagues provide a much needed roadmap for how to best approach proximity labeling experiments in C. elegans that will likely apply to other model systems.
They establish a rigorous approach by choosing to endogenously tag components of two essential NEKL-MLT complexes required for C. elegans molting. These complexes are relatively low abundance as they are only expressed in a single cell type, the hyp7 epidermal syncytium. In addition, as inactivation of any member of the complexes results in molting defects, they have a powerful selection for functional tags. Thus, they have set a high bar for themselves in order to discern whether a given variation on the experimental approach results in improved detection of interactors and fewer false positives.
Potential areas for improvement include lowering the expression level of the skin-specific soluble TurboID used to determine non-specific biotinylation events. This control results in much higher levels of biotinylation compared to the TurboID-tagged NEKL-MLT alleles and likely affects their analysis, which they openly admit. In addition, to reduce the high level of background biotinylation signals generated by endogenous carboxylases, they adopt a depletion strategy pioneered by other researchers but this does not offer major improvements in detection of specific signals. The source of these conflicting results remains to be determined. It is also curious that auxin-inducible degradation of components of the NEKL-MLT complexes did not robustly alter the resulting biotinylating capacity of other members. This approach should be evaluated in subsequent studies. Finally, as mentioned in Major Comment #3 (above), it would be interesting to see if combining TurboID tags within the same complex might improve signal-to-background ratios.
This manuscript represents a methodological advance that will likely become an oft-cited reference for members of the C. elegans community and a springboard for other basic biomedical scientists wanting to adapt rigorous proximity labeling techniques to their system. I am a cell biologist that uses a variety of genetic, molecular and biochemical approaches, mostly centered around C. elegans. I have used LC/MS-MS in our studies but have relatively little expertise in evaluating all aspects of proteomic pipelines.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This study expanded the use of data-independent acquisition-mass spectrometry (DIA-MS) in TurboID proximity-labeling proteomics to identify novel interactors of NEKL-2, NEKL-3, MLT-2, MLT-3, and MLT-4 complexes in C. elegans. The authors described several useful metrics to evaluate the quality of TurboID experiments, such as using the percentage of upregulated genes, the percentage of proteins present only in bait-TurboID experiments as compared to N2 controls, and the percentage of endogenously biotinylated carboxylases as internal controls. Further, the authors introduced methodological variability across 23 TurboID experiments and evaluated any improvement to the resulting data, such as N-terminally tagging bait proteins with TurboID, depleting endogenous carboxylases, and auxin-inducible degradation of known complex members. Finally, this study identified the kinase folding chaperone CDC-37 and the WASH complex component DDL-2 as novel interactors with the NEKL-MLT complexes through an RNAi-based enhancer approach following their identification by TurboID.
Major comments:
The key conclusions are convincing, and the work is rigorous. The work provides a clear roadmap to reproducing the data. The experiments are adequately replicated, and statistical analysis is adequate. We only have minor comments.
Minor comments:
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Proximity labeling has become a powerful tool for defining protein interaction networks and has been utilized in a growing number of multicellular model systems. However, while such an approach can efficiently generate a list of potential interactors, knowledge of the most appropriate controls and standardized metrics to judge the quality of the data are lacking. The study by Fay systematically investigates these questions using the C. elegans NIMA kinase family members NEKL-2 and NEKL-2 and their known binding partners MLT-2, MLT-3 and MLT-4. The authors perform eight TurboID experiments each with multiple NEKL and MLT proteins and explore general metrics for assessing experimental outcomes as well as how each of the individual metrics correlates with one another. They also compare technical and biological replicates, explore strategies for identifying false positives and investigate a number of variations in the experimental approach, such as the use of N- versus C-terminal tags, depletion of endogenous biotinylated proteins, combining auxin-inducible degradation, and the use of gene ontology analysis to identify physiological interactors. Finally, the authors validate their findings by demonstrating that a number of the candidate identified functionally interact with NEKL-2 or components of the WASH complex.
Overall this is an outstanding study that will be of great interest to those interested in using proximity labeling to identify interactors of their favorite protein. The experiments are well executed and the data presented in a mostly clear manner. I really like this study (particularly because I plan to do a proximity labeling study of my own), but I did come away less than impressed with some of the analysis. This is a data-dense manuscript, and it appears to me that the authors tried to cover so much ground that in some cases very little insight was provided. For instance, the authors promote the use of data independent acquisition (DIA) as compared to the more commonly used data dependent acquisition (DDA). However the authors do not provide any analysis to indicate one approach is better than the other. Likewise the combined use of auxin-induced degradation and proximity labeling is explored but there is very little to take away from these experiments. Despite these issues, I am very enthusiastic about the study as a whole. Below I list major and minor concerns.
Major concerns
Minor concerns:
Line 92 typo. I believe the authors meant to say NEKL-2-MLT-2-MLT-4.
Line169. Is exogenous the correct word to use here? It suggests that you are talking about non-worm proteins, but I know you are not.
Line 177 typo (D) should be (C).
Figure 1C: Lucky Charms may sue you for infringement of their trademarked marshmallow treats.
Figure 1D The NEKL-2::TurboID band is indicated with a green triangle in the figure but the figure legend states that green triangles indicate mNG::TurboID control. I know this triangle is a shade off the triangle that indicates mNG::TurboID but it's really hard to see the difference. All of the differently colored triangles in panel F are unnecessary. I would either just pick one color for all non-control bait proteins or better yet, only use a triangle to point to bands that are not obvious. For instance I don't need the triangles that point to NEKL-2 -3 and -4 fusion proteins. These are just distracting.
Line: 316: Conceivably, another factor that could contribute to the counterintuitive upregulation of some proteins in the N2 samples is related to the fusion proteins that are being expressed in the TurboID lines. A partially functional bait protein (one with a level of activity similar to nekl-2(fd81) that may not result in an obvious phenotype) could directly or indirectly affect gene expression leading to lower levels of a subset of proteins in the TurboID samples. The same could be said for fusion proteins with a gain-of-function effect.
Fig 3 B-E. I am a little confused how the data in these graphs is normalized. For instance, I would have expected that for NEKL-3 in panel B, that the normalized (log2) intensity value in N2 be set at 0 as it is for NEKL-2. Maybe I just don't have enough information on how these plots were generated.
Figure 6C legend is not correct.
Line 575: Figure reference should be Fig. S5G. The authors should check to make sure all references to supplemental figures include correct panel information.
Line 576. The authors reference a study by Artan and colleagues and report a weak correlation between their study and that of Artan. They reference figure S4 but it should be Fig S5H.
Line 652. The authors note that numerous proteins were present at substantially reduced levels in the mNG::TurboID samples and suggest that sticky proteins may have been outcompeted or otherwise excluded from beads incubated with the mNG::TurboID lysates. Why would sticky proteins only be a problem in these samples? The reasoning is not clear to me.
Line 745: The term "bait overlaps" is a bit vague. Ultimately, I figured out what it meant but it was not immediately obvious.
S7B Fig. Why is actin missing from the eluate?
Line 873: The authors state the extent of overlap in GO terms between the various experiments and provide percentages. I tried to extract this information from Figure 8C and came up with different values. For instance, in the case of Molecular Function, they state that they observed a 54% overlap between NEKL-2 and NEKL-3 but in the Venn diagram in Figure 8C I see that the NEKL-2 and NEKL-3 experiments had 71 (25+46) GO terms in common. Out of 98 GO terms for NEKL-2 or 104 for NEKL-3 the percentage I got is closer to 72. Am I analyzing this correctly?
Overall this is an outstanding study that will be of great interest to those interested in using proximity labeling to identify interactors of their favorite protein. The experiments are well executed and the data presented in a mostly clear manner. I really like this study (particularly because I plan to do a proximity labeling study of my own), but I did come away less than impressed with some of the analysis. This is a data-dense manuscript, and it appears to me that the authors tried to cover so much ground that in some cases very little insight was provided. For instance, the authors promote the use of data independent acquisition (DIA) as compared to the more commonly used data dependent acquisition (DDA). However the authors do not provide any analysis to indicate one approach is better than the other. Likewise the combined use of auxin-induced degradation and proximity labeling is explored but there is very little to take away from these experiments. Despite these issues, I am very enthusiastic about the study as a whole.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
The authors do not wish to provide a response at this time
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary: The authors present ASPEN - a tool for allelic imbalance estimation in haplotype-resolved single-cell RNA-seq data. Besides the mean of the allelic ratio, ASPEN manages to assess its under- and overdispersion as well as perform group-level comparisons. Dr. Wong with colleagues applied ASPEN to the simulated and publicly available single-cell data from mouse brain organoids and T cells. They showed a general applicability of the tool to this type of data, compared it with scDALI in terms of statistical power, and made numerous conclusions regarding the allele-specific regulation of housekeeping and cell-specific gene expression in general and during cell differentiation, as well as identified examples of X inactivation, imprinting and random monoallelic expression.
Major comments:
Minor comments:
Nowadays the allele-specific gene expression analysis using single-cell RNA-seq data is widely used to study allele-specific bursting [https://doi.org/10.1186/s13059-017-1200-8], imprinting, X chromosome inactivation [https://doi.org/10.1038/s42003-022-03087-4] and other processes [https://doi.org/10.1016/j.tig.2024.07.003].
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The authors introduce ASPEN (Allele-Specific Parameter Estimation in scRNA-seq), a statistical framework designed to model cis-regulatory variation in single-cell RNA-sequencing data, and demonstrate that ASPEN effectively detects cell state-specific allelic imbalances. Using simulated datasets, the authors show that ASPEN outperforms existing methods (e.g., scDALI) in both sensitivity and specificity. Furthermore, they demonstrate that ASPEN can be used to further dissect allelic imbalance, enabling the identification of random monoallelic expression (RME), gene expression pulsing, and dynamic regulatory shifts.
My main concerns are:
The ASPEN framework is useful for identifying single cell ASE and related analysis, which currently is under developed. It is timely and the framework is rigorous and flexible and driving by the data.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This is an interesting paper, which introduces a new approach and software ASPEN for analysis of allele-specific gene expression, which is applied to transcriptomes of F1 hybrids of mouse lines. The manuscript introduces an interesting statistical technique, which up to my knowledge is correct and brings about new biological results, identifying genes with systematically decreased or increased expression variance and statle allelic expression ratio, which seems to be controlled by the regulatory machinery.
The manuscript has some shortcomings in presentation, it is written very concisely, especially in its methods part, and is somewhat difficult to follow.
I'm not sure that the authors make the correct claim in the manuscript. The title and the abstract says that the manuscript discusses the cis-regulatory heterogeneity, but in fact there is very little in the manuscript about gene regulation per ce. The study demonstrates that allele specific expression is controlled by some yet unknown mechanisms, rather than a product of technical noise and then presents a number of examples of different pathways which the increased and decreased allele specific variance. Also the manuscript presents several examples of shifts in the variance of particular genes in temporal development.
Yet, the manuscript tells virtually nothing about regulation, thus the conclusion that 'ASPEN enables the interrogation of cis regulatory effects on gene expression' is not justified in its literal terms; what ASPEN does it quantifies the allele-specific transcription activity effects in a single cell transcriptomics experiment. Mechanistically the observed effects can be explained by any regulatory effect like DNA methylation, chromatin structure or whatever. To prove that cis-regulatory effects are important here the authors need to show the allele specific nature of transcription factor binding (for instance by showing the TF binding motifs destroyed/created by variants). It is more difficult to take into account the chromatin effects without ATAC experiments but it might be that ATAC-seq experiments are available for parental line and there is a differential DNA accessibility in the locality of genes of interest. I think only with such mechanistic illustrations one can conclude that cis-regulatory interactions play a major role here.
As an other option, the authors may publish the study per se but with a changed title, the abstract and the discussion, formulating it in a more phenomenological way.
Minor note
In Figures 2-5 the low variance genes are shown with dots occupying lines parallele to x axis. This can be related to some wrong digitising of variance or to a low numbers of reads contributing to the variance. Please double check.
The paper introduces a new interesting statistical approach for quantifying allele specific transcription from the single cell data, using Bayesian shrinkage technique similar to that used in edgeR. The paper has clear biological meaning demonstrating that there are genes with a decreased variability in gene expression. I believe, the paper draws attention to the interesting area of facts and as such may be published.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Manuscript number: RC-2025-03031
Corresponding author(s): Lara-Pezzi, Enrique and Gómez-Gaviro, María Victoria
Following the review of our article entitled "Loss of the alternative calcineurin variant CnAβ1 enhances brown adipocyte differentiation and drives metabolic overactivation through FoxO1 activation", we propose below a number of experiments to be performed in order to address the issues raised by the reviewers.
While we acknowledge the limitations of the full CnAβ1 knockout mouse and we unfortunately lack a tissue-specific knockout mouse, we believe that the proposed new experiments together with the (abundant) existing information in the paper will help clarify the concerns raised by the reviewers.
Insert here a point-by-point reply that explains what revisions, additional experimentations and analyses are planned to address the points raised by the referees.
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
*The current study examines the metabolic phenotype of mice lack the calcineurin variant CnAb1 (CnAb1KO). On a high fat diet, CnAb1KO mice gain less weight compared to WT controls, which is accompanied by improvements in obesity-related metabolic dysfunction, such as glucose/insulin intolerance and hyperlipidemia. The authors attribute most of the observed phenotypes to enhanced brown fat function, notably fatty acid catabolism and the thermogenic capacity. Mechanistically, the authors propose that CnAb1KO increases FoxO1 transcriptional activity, as a result of reduced mTOR/Akt signaling, which in turn mediates the hyper-catabolism of BAT in CnAb1KO mice. *
* Major comments: *
*Q1. The main issue of the study is it's not hypothesis driven. Based on high fat diet-induced metabolic phenotype of the whole body CnAb1KO mice, the authors put together a mechanism focusing on potential roles of CnAb1 in BAT functions that affect systemic metabolic homeostasis. However, the rationales to establish this link were based largely on correlative results and at times incorrect data interpretation (for instance, using the expression of Myf5 and Pax7 as markers for brown adipocyte differentiation). The sequential event from CnAb1 loss of function to reduced mTOR signaling and increased FoxO1 activity (or conversely, how CnAb1 increases mTOR signaling to reduce FoxO1 activity) has not been mechanistically characterized. There are also no studies to explain how FoxO1 is involved in brown fat differentiation and hyper-catabolism of BAT downstream of the CnAb1-mTOR pathway. In addition, the UCP-1 FoxO1KO experiment in Fig. 6 fails to provide strong evidence to support the claim. Thus, there are many gaps between the observed phenotype and the proposed mechanism. *
A1. We thank the reviewer for the insightful comments. We agree with the reviewer that, historically, this project did not originally focus on the BAT. Instead, we arrived at the BAT after ruling out other possibilities to explain the reduced body weight observed in these animals, together with the reduced body temperature after starvation, which was our first observation. While the BAT involvement was not our first hypothesis a priori, we do not agree that this would invalidate or reduce the interest of our work. While our initial evidence may have been correlative at first, the FoxO1 BAT-specific knockout experiments and the AAV/Ucp1-Cre CnAβ1 expression restoration experiments prove that the BAT is indeed involved in the phenotype observed in CnAβ1Δi12 (KO) mice. It is likely that other organs may be also involved (since the phenotype is not fully prevented by the BAT-specific approaches) but the BAT is definitely involved.
To further substantiate the involvement of the BAT in the improved metabolic phenotype observed in CnAβ1Δi12 mice, we propose to perform BAT transplantation, monitoring body weight over 8 weeks following transplantation. If successful, BAT transplantation from CnAβ1Δi12 mice into WT mice should improve their metabolic response to high-fat diet (HFD), thereby reinforcing the role of the BAT in these mice.
In addition, we propose to measure the __*levels of so-called batokines*__ FGF21, VEGFA, IL6, and also of 12,13-diHOME in BAT and serum from 12-week-old chow and HFD mice.
With regards to Pax7 and Myf5, while we agree that these are common precursors to other lineages (skeletal muscle), we show in Fig. S1E additional differentiation markers such as Cox2 and Cpt1b. __*The 5 markers assessed showed an increase in *____*CnAβ1Δi12 mice, pointing towards a cell-autonomous effect of the absence of CnAβ1 on the BAT*__. Nevertheless, to further substantiate the accelerated differentiation of brown preadipocytes in the absence of CnAβ1, we propose to __*measure the expression of additional BAT markers*__ (although they are not exclusive of BAT), such as Ucp1, Prdm16, PPARγ, and AdipoQ in brown preadipocytes isolated from 6–8-week-old mice.
With regards to the activation of mTOR (specifically mTORC2) by CnAβ1, we published this in previous papers from our group: Gómez-Salinero et al (Cell Chem Biol, 2016), Felkin et al (Circulation, 2011), Lara-Pezzi et al (J Cell Biol 2007), Padrón-Barthe et al (J Am Coll Cardiol 2018). The mechanism involves the interaction between CnAβ1 and mTORC2 in cellular membranes. Knockdown of CnAβ1 results in mTORC2 mislocalisation and Akt inhibition. In addition, we show in Fig. 6C in this paper that PTEN inhibition reduces the improved differentiation of BAT adipocytes from CnAβ1Δi12 mice, further involving the Akt pathway in the observed phenotype. Furthermore, Fig. 6 shows a significant increase in body weight and BAT weight in BAT-specific FoxO1 knockout CnAβ1Δi12 mice, together with a significant decrease in different Pnpla1, Irf4, and Bcat2 expression. While we agree that the reversal of the phenotype is only partial, the effect of knocking out FoxO1 in the BAT of CnAβ1Δi12 mice is both statistically significant and biologically relevant. We would be happy to provide additional information at the Editors’ request. In addition, we propose to carry out __BAT preadipocyte differentiation experiments comparing cells isolated from CnAβ1Δi12 mice to those isolated from CnAβ1Δi12 mice with BAT-specific FoxO1 knockout__.
Q2. A second issue is that most of the phenotypes can be explained by the difference in weight gain. With the available data, it's difficult to pinpoint the tissue origin(s) mediating the weight gain/loss phenotype. The authors would first need to generate a BAT-CnAb1KO mouse line to convincingly show a main role for BAT CnAb1 in systemic metabolic homeostasis. There are also many problems with data presentations/interpretations of the metabolic phenotyping studies. For example, Fig. 1A shows that CnAb1KO mice are about 5 g lighter than controls. However, Fig. 1G indicates a 10 g difference in fat mass. The EM images in Fig. 3B are of poor quality, which seems to suggest that HFD fed CnAb1KO mice have the highest mitochondrial density. Lastly, in Fig. 4C/D, the authors interpret the reduced FFA and glycerol levels in CnAb1KO after b3-agonist injection as increased fatty acid burning by BAT, which is incorrect. If anything, the reduced glycerol release in the KO mice would suggest a reduction in lipolysis. However, the most likely explanation is that WT mice have more fat mass and as such, more fat hydrolysis.
A2. While we agree with the reviewer that some of the features may be explained by reduced body weight gain (reduced WAT weight, for instance), many other changes showed by CnAβ1Δi12 mice cannot be explained by reduced body weight gain alone, including higher expression of differentiation markers in BAT, higher number of mitochondria in BAT, or improved cold-tolerance, among others. Therefore, we respectfully disagree with the reviewer’s opinion.
Unfortunately, we do not have a tissue-specific CnAβ1 knockout mouse and we cannot commit to having one in the short term. While we acknowledge the limitations of using a full knockout mouse, we provided several pieces of evidence that the BAT is involved in the observed phenotype, as pointed out in the discussion: 1) Placing CnAβ1Δi12 mice in thermoneutral conditions mitigated the weight loss. 2) Reintroducing CnAβ1 in BAT with a CnAβ1-overexpressing virus partially prevented the weight loss. 3) Minimal changes in mitochondrial gene expression were observed in skeletal muscle and liver, suggesting that the phenotype is primarily driven by alterations in BAT. 4) BAT adipocytes from CnAβ1Δi12 mice differentiated more effectively than those from wild type mice, suggesting a cell-autonomous effect. While a direct effect of CnAβ1 on WAT cannot be entirely ruled out, our results strongly suggest that loss of CnAβ1 in BAT is a major contributor to the observed metabolic changes.
With regards to Fig. 1E, this is an estimation of fat weight from __MRI__ images. We agree with the reviewer that this is obviously wrong and we will __revise this quantification__. We propose to __add measurements of subcutaneous WAT__, which we also have, to further support the difference observed in eWAT.
With regards to Fig. 3B, we agree that some of the individual figures may have been poorly chosen, but the graph in Fig. 3C (which quantifies the electron microscopy pictures) clearly shows that the reduction in mitochondria in WT mice as a result of HFD feeding is prevented in CnAβ1Δi12 mice. Fig. 3C does not show an increase in mitochondria with HFD, as implied by the reviewer based on Fig. 3B. We propose to __provide adequate panels for Fig. 3B that better reflect the averages shown in Fig. 3C__.
Regarding Fig. 4C and D, we thank the reviewer for this correction, which we agree with. We still believe that the BAT of CnAβ1Δi12 mice is burning fat more effectively than that of WT mice, but we agree that these experiments are not the proof of this claim. We will__ move or remove panels C and D from Fig. 4__ and focus this figure on thermogenic capacity.
To assess systemic lipolysis, we will __measure in vivo serum levels of NEFA__ (non-esterified fatty acids) __and glycerol__ in 12-week-old mice fed a HFD. Additionally, to evaluate BAT lipolytic activation, we will perform __BAT explant and *ex vivo* experiments__ to determine the lipolysis rate. This should provide valuable information supporting the role of the BAT in the observed phenotype in CnAβ1Δi12 mice.
*Q3. The authors should take a fresh, unbiased look at existing data, form a testable hypothesis and design a series of new experiments (including new tissue-specific KO mice) to assess the function of CnAb1 in BAT or other tissues responsible for the metabolic phenotype. If BAT is indeed involved, the authors need to mechanistically determine the role of CnAb1 in brown adipocyte differentiation vs BAT function and explain why the ratio of CnAb1/CnAb2 ratio matters in this context, as this is the basis for the entire study. A revision addressing main issues of the manuscript will not likely to be completed in a typical revision time (e.g. 3 months). *
A3. As explained above, unfortunately we do not have tissue-specific CnAβ1 knockout mice. If the Editors consider that this is essential for resubmission of a revised article, we are afraid that we cannot comply. This said, we believe that our manuscript contains relevant data about metabolic regulation by the CnAβ1 calcineurin isoform that are new and relevant to the field.
Our data provide clear evidence that the BAT is indeed involved in the phenotype observed in CnAβ1Δi12 mice, as explained in our previous answers above. It may not be the *only* tissue involved, but it is most definitely involved. The BAT transplant experiments will add further evidence of this.
We already show evidence of the role of CnAβ1 (or rather, its absence) in the differentiation of BAT pre-adipocytes (Fig. S1E and Fig. 6C) and we will __provide additional evidence through the proposed new experiments__. Similarly, we provide evidence of the role of CnAβ1 in BAT weight, transcriptional profile, lipid content, and number of mitochondria. Also here, we believe that __the proposed experiments will reinforce this aspect of the paper__.
Reviewer #1 (Significance (Required)):
*Q4. The thermogenic capacity of brown and beige adipocytes has shown promise as a means to reduce fat burden to treat obesity and related metabolic diseases. Identification of brown/beige adipocyte promoting mechanisms may provide druggable targets for therapeutic development. As such, the topic and findings of the current study would be of interest to researchers in the metabolism and drug development fields. The weakness of the study is that it's descriptive and the authors jump to conclusions without strong supporting evidence. Most of the metabolic phenotypes associated with CnAb1KO mice are likely secondary to the weight difference. The rationale to focus on BAT is not well justified. A well-thought-out approach would be needed to identify the tissue origins mediating the metabolic phenotypes of CnAb1KO mice and to dissect the underlying mechanisms. *
*Reviewer's field of expertise: adipose tissue biology, systemic metabolic regulation, immunometabolism *
A4. We agree with the reviewer about the potential relevance of our findings. The shortcomings pointed out in this comment have been addressed above. Overall, we thank the reviewer for their thorough review of our ms.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
*The manuscript entitled « Loss of the alternative calcineurin variant CnAβ1 enhances brown adipocyte differentiation and drives metabolic overactivation through FoxO1 activation » by Dr Lara-Pezzi and colleagues describes the role of the calcium/calmodulin dependent serine/threonine phosphatase catalytic subunit calcineurin variant CnAß1 in brown adipose tissue physiology and function. Through the use of global CnAß1 KO mice, the authors show that these mice are resistant to diet-induced obesity, have increased thermogenesis due to increased mitochondrial activity, decreased body weight, improved glucose homeostasis, increased fatty acid oxidation. The authors also demonstrate that these effect are mostly mediated through improved brown adipose tissue (BAT) function, through increased Foxo1 activation in BAT. Genetic deletion of Foxo1 in BAT resulted in increased body weight and impaired mitochondrial gene expression. In addition, the authors also correlate their findings to potential CNAß1 polymorphism from the UK biobank associated to improved metabolic traits in humans (blood glucose mainly). *
Although interesting, the conclusion are not always supported by the data. The manuscript requires additional experiments to further consolidate their claims.
*Q1. It should be mentioned that all experiments are performed in global CnAβ1 KO mice. Thus, it is difficult to assess the cell-autonomous role if this protein in BAT function (even if an AAV9 driving CnAβ1 expression is used; or if other tissues have been studied). This should be discussed at least as a limitation of the study, except if floxed mice are available. *
A1. We thank the reviewer for the positive comments about our work.
Unfortunately, we do not have a tissue-specific CnAβ1 knockout mouse. However, we believe we provide abundant evidence of the involvement of the BAT in the phenotype observed in CnAβ1Δi12 mice, including the following: 1) Placing CnAβ1Δi12 mice in thermoneutral conditions mitigated the weight loss. 2) Reintroducing CnAβ1 in BAT with a CnAβ1-overexpressing virus partially prevented the weight loss. 3) Minimal changes in mitochondrial gene expression were observed in skeletal muscle and liver, suggesting that the phenotype is primarily driven by alterations in BAT. 4) BAT adipocytes from CnAβ1Δi12 mice differentiated more effectively than those from wild type mice, suggesting a cell-autonomous effect. While a direct effect of CnAβ1 on WAT cannot be entirely ruled out, our results strongly suggest that loss of CnAβ1 in BAT is a major contributor to the observed metabolic changes.
This said, we fully agree with the reviewer to acknowledge in the discussion the limitation of using a full knockout mouse for this study.
Q2. Is there good antibodies for CnAβ1? The protein levels of the protein should be shown in, at least, adipose tissues of WT and KO mice under chow and HFD.
A2. There is no good antibody against CnAβ1. The main reason is that the C-ter domain of this isoform is not very immunogenic. We did try to generate an antibody, but we got no immune response against the unique C-ter domain. We do have an old antibody generated against CnAβ1 years ago. We propose to try to perform WB and immunohistochemistry in WT and ____CnAβ1Δi12 mice. However, we need to be clear that we cannot make any commitments towards these results, since the antibody may not work. In any case, we believe that the RT-PCR results, which clearly discriminate both isoforms, are very clear.
*Q3. A general comment is that most of the conclusions are drawn from qRT-PCR data. It lacks functional experiments that may reinforce the conclusion. For example, did the authors measure mitochondrial function in BAT of WT and KO mice using different substrate (fatty acids, glucose, ...)? *
A3. We thank the reviewer for this suggestion and we therefore propose to include in the revised paper measurements of mitochondrial activity with different substrates in WT and ____CnAβ1Δi12 mice.
*Q4. Lack of validation of the mouse model used (CnAβ1 expression in BAT upon AAV9 over expression confirmed? What about the other tissues?). *
A4. We showed in Fig. 5E the increase in CnAβ1 expression in the BAT of Ucp1-Cre mice infected with the floxed AAV-CnAβ1 virus. We propose to include similar expression analyses in other tissues.
Reviewer #2 (Significance (Required)):
Q5. This is a novel study addressing the role of CnAβ1 in energy homeostasis, more specifically in BAT function. This study reports for the first time the role of CnAβ1 in energy homeostasis, with new mechanistic insights related to the crosstalk between CnAβ1 and Foxo1.
The authors have previously described the role of this protein in cardiac function. There are not a lot of publications describing the function of this protein, thus this study may be interested for the community working on diabetes/obesity/cardio-metabolic field.
*Limitations : see below (lack of functional data, ...). *
A5. We thank the reviewer for these comments, with which we agree.
*
*
As much as we would like to have a tissue-specific CnAβ1 knockout mouse, the reality is that we do not have it. In any case, we believe that our paper provides a considerable amount of data that is relevant to the field.
We remain open to incorporating the suggested experiments, or others, should they be considered necessary to further strengthen the manuscript.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript entitled « Loss of the alternative calcineurin variant CnAβ1 enhances brown adipocyte differentiation and drives metabolic overactivation through FoxO1 activation » by Dr Lara-Pezzi and colleagues describes the role of the calcium/calmodulin dependent serine/threonine phosphatase catalytic subunit calcineurin variant CnAß1 in brown adipose tissue physiology and function. Through the use of global CnAß1 KO mice, the authors show that these mice are resistant to diet-induced obesity, have increased thermogenesis due to increased mitochondrial activity, decreased body weight, improved glucose homeostasis, increased fatty acid oxidation. The authors also demonstrate that these effect are mostly mediated through improved brown adipose tissue (BAT) function, through increased Foxo1 activation in BAT. Genetic deletion of Foxo1 in BAT resulted in increased body weight and impaired mitochondrial gene expression. In addition, the authors also correlate their findings to potential CNAß1 polymorphism from the UK biobank associated to improved metabolic traits in humans (blood glucose mainly).
Although interesting, the conclusion are not always supported by the data. The manuscript requires additional experiments to further consolidate their claims.
It should be mentioned that all experiments are performed in global CnAβ1 KO mice. Thus, it is difficult to assess the cell-autonomous role if this protein in BAT function (even if an AAV9 driving CnAβ1 expression is used; or if other tissues have been studied). This should be discussed at least as a limitation of the study, except if floxed mice are available. Is there good antibodies for CnAβ1? The protein levels of the protein should be shown in, at least, adipose tissues of WT and KO mice under chow and HFD.
A general comment is that most of the conclusions are drawn from qRT-PCR data. It lacks functional experiments that may reinforce the conclusion. For example, did the authors measure mitochondrial function in BAT of WT and KO mice using different substrate (fatty acids, glucose, ...) ? Lack of validation of the mouse model used (CnAβ1 expression in BAT upon AAV9 over expression confirmed? What about the other tissues?).
This is a novel study addressing the role of CnAβ1 in energy homeostasis, more specifically in BAT function. This study reports for the first time the role of CnAβ1 in energy homeostasis, with new mechanistic insights related to the crosstalk between CnAβ1 and Foxo1.
The authors have previously described the role of this protein in cardiac function. There are not a lot of publications describing the function of this protein, thus this study may be interested for the community working on diabetes/obesity/cardio-metabolic field.
Limitations: see below (lack of functional data, ...).
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The current study examines the metabolic phenotype of mice lack the calcineurin variant CnAb1 (CnAb1KO). On a high fat diet, CnAb1KO mice gain less weight compared to WT controls, which is accompanied by improvements in obesity-related metabolic dysfunction, such as glucose/insulin intolerance and hyperlipidemia. The authors attribute most of the observed phenotypes to enhanced brown fat function, notably fatty acid catabolism and the thermogenic capacity. Mechanistically, the authors propose that CnAb1KO increases FoxO1 transcriptional activity, as a result of reduced mTOR/Akt signaling, which in turn mediates the hyper-catabolism of BAT in CnAb1KO mice.
Major comments:
The thermogenic capacity of brown and beige adipocytes has shown promise as a means to reduce fat burden to treat obesity and related metabolic diseases. Identification of brown/beige adipocyte promoting mechanisms may provide druggable targets for therapeutic development. As such, the topic and findings of the current study would be of interest to researchers in the metabolism and drug development fields. The weakness of the study is that it's descriptive and the authors jump to conclusions without strong supporting evidence. Most of the metabolic phenotypes associated with CnAb1KO mice are likely secondary to the weight difference. The rationale to focus on BAT is not well justified. A well-thought-out approach would be needed to identify the tissue origins mediating the metabolic phenotypes of CnAb1KO mice and to dissect the underlying mechanisms.
Reviewer's field of expertise: adipose tissue biology, systemic metabolic regulation, immunometabolism
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
REVIEWER 1
This is an important and solid study that identified sequences that can improve circRNA translation and that as or more importantly are very short and hence are suitable for generating of efficient protein expressing circRNAs. This manuscript fills an important gap in the field, and it is highly significant. The study is well controlled, the rationale clear and the results conclusive with no major flaws.
While this is a minor concern as the vector has been used before, it will greatly improve the quality of the paper if the authors could just verify that the vector only generates circRNA molecules and not linear concatenamers. To do so the authors can focus only in their control and the most optimal transcripts and perform northern blot or well controlled RNAseR experiments to show that all RNA molecules containing the back splicing junction are circular We thank the reviewer for raising this point. As suggested, we performed RNaseR resistance assays on our three most efficient candidates driving cGFP translation (VCIP, T3-glo, and T3-U3) to confirm that all derived RNA molecules containing the back-splicing junction are circular. As proof of this, cGFP proved strongly resistant to RNase R (new Fig. S1N), confirming its circular structure. We further ruled out the possibility that molecules other than the circRNA encoding GFP serve as templates for translation from our vectors. Specifically, ad hoc PCR amplifications performed for this purpose (new Fig. S1M) showed no bands that would indicate the presence of concatemers. Indeed, ad hoc PCR amplifications (new Fig. S1M) revealed no bands indicative of concatemer formation. The primers used and the expected sizes of the amplicons are schematically represented in new Fig. S1M. In brief, we used a divergent primers set spanning the BSJ (3-4) to specifically detect the mature circRNA and a set of convergent primers (1-2) pairing on the GFP ORF, thus detecting both the circRNA and its linear precursor as well as the putative concatemer expected. Although a ~1 kb band was expected if a trans-splicing by-product was present, no such band was observed (new Fig. S1M). Moreover, RT-PCR amplification of the cGFP back-splice junction was markedly more efficient when reverse transcription was primed with random hexamers than with oligo(dT), priming total RNA or preferentially polyA+ RNA, respectively. These results are expected for a circRNA, as also indicated by the fact that the circZNF609 positive control behaves in a similar manner. Collectively, these results confirm the circular nature of our transcript and exclude translation originating from possible concatemers.
These results are shown in new Fig. S1M and S1N and described in the text as follows: “Importantly, we ruled out the possibility that templates other than the GFP-encoding circRNA drive translation from our best performing constructs (V-cGFP, T3-glo-cGFP and T3-U3-cGFP). Ad hoc PCRs amplifications (Fig. S1M) revealed no bands indicative of concatemer formation. The left panel of Fig. S1M schematically illustrates the primer sets and expected amplicons sizes. In particular, we used a divergent primers set spanning the BSJ (3-4) to specifically detect the mature circRNA and a set of convergent primers (1-2) pairing on the GFP ORF detecting both the circRNA and its linear precursor as well as the putative concatemer expected. Although a ~1 kb band was expected if a trans-splicing by-product was present, no such band was observed. Moreover, RT-PCR amplification of the cGFP back-splice junction was markedly more efficient when reverse transcription was primed with random hexamers than with oligo(dT), priming total RNA or preferentially polyA+ RNA, respectively (Fig. S1M). These results are consistent with the circularity of the transcripts tested and coherent with the results obtained for circZNF609, used as control (Fig. S1M). Finally, cGFP resulted resistant to RNAseR treatment (Fig. S1N), further supporting its circular nature.”*
There is a repetition of the world "a" in the abstract. We thank the reviewer for the attention paid to our text, we removed the extra “a” from the abstract.
All circRNA translation studies should be cited when describing translation of circRNAs. We thank the reviewer for the suggestions, we corrected the mistake present in the text and included extra referenced about circRNA translation.
*Specifically, we included: *
Circular RNAs (circRNAs) have attracted significant interest due to their unique properties, which make them promising tools for expressing exogenous proteins of therapeutic value. However, several limitations must be addressed before circRNAscan become a biologically and economically viable platform for the biotech industry.One of the main challenges is the reliance on large, highly structured sequences withinternal ribosome entry site (IRES) activity to initiate translation of the downstream open reading frame. In this study, the authors propose an alternative strategy that combines the 5′ untranslated region (5′UTR) of a previously characterized natural circRNA(circZNF609) with a short 13-nt nucleotide sequence shown to act as a translational enhancer. By evaluating the activity of various constructs containing a reporter geneacross multiple cell lines, they identify the most efficient and compact sequence, 63-nt long, capable of boosting translation within a circular RNA context.
Major Comments:
This study is well-executed and relies on standard in vitro molecular biology techniques, which are adequate to support the conclusions drawn. *We thank the reviewer for the very positive opinion on the execution of our study. *
The experimental procedures are clearly described, and the statistical analyses have been performed according to accepted standards. *We thank the reviewer for the very positive comment about the analyses we performed. *
Minor Comments:
The manuscript would greatly benefit from a comprehensive revision to improve clarity and language. Involving a native English speaker during the editing process could significantly enhance the manuscript's readability and overall quality. The Results section would benefi t from closer attention, as certain parts of the description are attimes confusing and could be clarifi ed for better reader comprehension. We thank the reviewer for the input. We performed a huge revision of the text to improve language quality and enhance readability. We extended the descriptions in the results sections in order to explicit and clarify our data.
The references should be carefully reviewed for accuracy and consistency-forinstance, references 9 and 10 appear to require correction or clarifi cation. We thank the reviewer for the careful reading of our paper. We amended the reference section, and we expanded it.
Reviewer #2 (Significance (Required)):
This study addresses a critical bottleneck in RNA therapeutics. The use of the proposed short sequences could significantly enhance the in vivo activity of protein-encoding circular RNAs. A highly efficient, compact translational enhancer has thepotential to substantially improve the therapeutic applicability of circRNAs and broaden their range of applications. Given the potential utility of these findings, we would anticipate pursuing intellectual property (IP) protection. To further strengthen the study, future work should include additional data on polysome association and a detailed analysis of the secondary structure of the 66-nt enhancer sequence. This work should be of broad interest to molecular biologists working on RNA biology, translation, and RNA-based therapeutics. I expect the identified sequence will betested by multiple laboratories to evaluate its strength and versatility, further underscoring the potential impact of this study. For context, I am actively engaged in research on non-coding RNAs.
REVIEWER 3
In this brief report, the authors take advantage of circular RNA expression plasmids to define elements that can be used to enable efficient translation. They test a handful of known IRES elements as well as short translation enhancing elements (TEEs) for their ability to promote translation of circular GFP and c-ZNF609 reporters. They focus on one particular element that is of a short length and seems to work as well as longer IRES elements. My major concern relates to possible alternative sources of the translated proteins, which the authors have not ruled out (see below). I find themanuscript to be too preliminary in its current state.
These results are shown in new Fig. S1M and S1N and described in the text as follows: “Importantly, we ruled out the possibility that templates other than the GFP-encoding circRNA drive translation from our top constructs (V-cGFP, T3-glo-cGFP and T3-U3-cGFP). Ad hoc PCRs amplifications (Fig. S1M) revealed no bands indicative of concatemer formation. The left panel of Fig. S1M schematically illustrates the primer sets and expected amplicons sizes. In brief, we used a divergent primers set spanning the BSJ (3-4) to specifically detect the mature circRNA and a set of convergent primers (1-2) pairing on the GFP ORF detecting both the circRNA and its linear precursor as well as the putative concatemer expected. Although a ~1 kb band was expected if a trans-splicing by-product was present, no such band was observed (new Fig. S1M). Moreover, RT-PCR amplification of the cGFP back-splice junction was markedly more efficient when reverse transcription was primed with random hexamers than with oligo(dT), priming total RNA or preferentially polyA+ RNA, respectively (Fig. S1M). These results are consistent with the circularity of the transcripts tested (Fig. S1M). Importantly, cGFP PCR amplifications showed similar results as a validated endogenous circRNA, namely circZNF609, used as control (Fig. S1M, right panel), confirming the circular nature of cGFP. Finally, cGFP resulted resistant to RNAseR treatment (Fig. S1N), further supporting its circular nature.”* *
Echoing the point above, the overall results would be stronger if the authors couldconfirm IRES activity using highly pure, in vitro transcribed RNAs that are transfected into cells * We thank the reviewer for this suggestion. Unfortunately, we are currently unable to produce synthetic circular molecules in-house, and the cost and time for purchasing synthetic ones are prohibitive. Nevertheless, we have performed the experiments described above to ensure the circularity of the transcripts tested.*
The authors should also confirm their IRES activity using standard dual luciferase reporter (linear) constructs which have long been a standard approach in the field. We thank the reviewer for raising this point. As recommended, we cloned our three best candidates (VCIP, T3-glo, and T3-U3) into the pRL-TK/pGL3 dual-luciferase vector to assess their IRES activity (producing the vectors VCIP-Luc, T3-glo-Luc, and T3-U3-Luc), transfected them into RD cells, and, after 24 h of incubation, measured luciferase activity to assess the IRES performance of each candidate. From our analyses, VCIP and T3-U3 confirmed their IRES activity, although showing different relative efficiency, whereas T3-glo was inactive in the linear luciferase context. This finding is consistent with previous observations (Legnini et al., 2017) showing that the performance of IRES sequences in a linear luciferase reporter may differ from their activity when driving translation from a circRNA template. Overall, these results highlight the need for further investigation into the sequences and contexts specifically governing circRNA translation, rather than relying solely on knowledge derived from linear RNAs. *The results are shown below. We did not include them in the text to not overcomplicate the readability. However, we are happy to add and discuss them if required. *
***
***
Bar plot representing the relative luciferase activity deriving from VCIP-Luc (“V”), T3-glo-Luc (“T3-glo”), and T3-U3-Luc (“T3-U3”)*. Dual luciferase assay was performed and Renilla luciferase activity from each candidate was normalized against the Firefly luciferase. An empty ptKRL-pgl3 vector was used as reference. The ratio of each sample versus its experimental control was tested by two-tailed Student’s t test. * indicates a Student’s t test-derived p-value * *
Methods, Plasmids Construction Section: Rather than including long lists of oligos and forcing a reader to figure out the final product that was cloned, it would be more intuitive if the authors provided the full sequences of the ORF and IRES sequencesthat were tested. We thank the reviewer for the comment, we added the sequences to the methods (Supplementary Table 1).
The manuscript needs extensive English editing. Parts of it are also formatted in anunusual style, especially the introduction where it seems like each paragraph is a single sentence. As requested by the reviewer, we edited the text to make the language and content more accessible to readers.
References included by the authors are selective and surprisingly do not include Chen et al (2021) Mol Cell 20:4300-4318 which already defined IRES elements for circRNAs that are fairly small. *Thank you for pointing this out. We have now cited the elegant work of Chen et al. (2021, Mol Cell 20:4300–4318) in the revised manuscript. While Chen and colleagues screened IRES-like elements of roughly 200 nt, our study was designed to uncover an even more minimal motif. The elements we report are therefore markedly shorter, highlighting a complementary, rather than overlapping, aspect of IRES available for driving circRNA translation. However, we now refer to Chen et al. in our text. *
Error bars in Fig 2, especially Fig 2B, are huge. It seems impossible to make any conclusion given the large variety across these experiments. Thank you for your input. Although the error bars appear relatively large, the overall conclusions remain robust, as also noted by the other reviewers: both T3-glo and T3-U3 are intrinsically compact elements, yet they drive translation as efficiently as larger canonical IRESs. The error bars largely reflect the inherent variability of transient transfection assays, which naturally increases with the number of constructs examined. To strengthen our dataset without discarding existing replicates, we chose not to repeat experiments in the previously tested lines. Instead, we assessed our vectors in an additional model, the D283 medulloblastoma cell line. In this setting, we unexpectedly observed that the EMCV IRES surpasses the VCIP IRES, opposite to what we saw in the other lines, yet even here the short elements we identified remain strong competitors (new Fig. 2C, S2G, S2H). The evaluation of multiple CDSs across several cell lines, make our findings to be solid and well supported.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this brief report, the authors take advantage of circular RNA expression plasmids to define elements that can be used to enable efficient translation. They test a handful of known IRES elements as well as short translation enhancing elements (TEEs) for their ability to promote translation of circular GFP and c-ZNF609 reporters. They focus on one particular element that is of a short length and seems to work as well as longer IRES elements. My major concern relates to possible alternative sources of the translated proteins, which the authors have not ruled out (see below). I find the manuscript to be too preliminary in its current state.
Major comments:
Minor comments:
Referees cross-commenting
I stand by my comments regarding the need for the authors to perform additional controls and validation.
This work is most relevant for researchers aiming to use circular RNAs as therapeutic modalities to express proteins. Defining optimal methods, including IRES elements, that enable maximal translational output would be helpful. Note, however, that this is far from the first study to look for IRES elements in circular RNAs (e.g. Chen et al (2021) Mol Cell 20:4300-4318) which did it in a much more extensive manner.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Circular RNAs (circRNAs) have attracted significant interest due to their unique properties, which make them promising tools for expressing exogenous proteins of therapeutic value. However, several limitations must be addressed before circRNAs can become a biologically and economically viable platform for the biotech industry. One of the main challenges is the reliance on large, highly structured sequences with internal ribosome entry site (IRES) activity to initiate translation of the downstream open reading frame. In this study, the authors propose an alternative strategy that combines the 5′ untranslated region (5′UTR) of a previously characterized natural circRNA (circZNF609) with a short 13-nt nucleotide sequence shown to act as a translational enhancer. By evaluating the activity of various constructs containing a reporter gene across multiple cell lines, they identify the most efficient and compact sequence, 63-nt long, capable of boosting translation within a circular RNA context.
Major Comments:
Minor Comments:
This study addresses a critical bottleneck in RNA therapeutics. The use of the proposed short sequences could significantly enhance the in vivo activity of protein-encoding circular RNAs. A highly efficient, compact translational enhancer has the potential to substantially improve the therapeutic applicability of circRNAs and broaden their range of applications.
Given the potential utility of these findings, we would anticipate pursuing intellectual property (IP) protection.
To further strengthen the study, future work should include additional data on polysome association and a detailed analysis of the secondary structure of the 66-nt enhancer sequence.
This work should be of broad interest to molecular biologists working on RNA biology, translation, and RNA-based therapeutics. I expect the identified sequence will be tested by multiple laboratories to evaluate its strength and versatility, further underscoring the potential impact of this study.
For context, I am actively engaged in research on non-coding RNAs.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In the manuscript entitled "A Short 63-Nucleotide Element Promotes Efficient circRNA Translation", Biagi et al. aim to identify sequences and layouts that would allow high expression of proteins from an engineered circular RNA (circRNA). Briefly, the authors utilize a circRNA-producing plasmid that produces a GFP protein encoded across the splice junction when translated and test different IRESs in combination with Translation Enhancing Element (TEEs). While performing these experiments they found that a short sequence containing the TEE (13-glo) is enough to promote significant levels of translation while keeping the size of the circRNA small. The authors then tested whether the presence of a spacer could help improving translation and identified a 50base sequence that in combination with the TEE can promote very efficient translation. The authors then went on and showed that this element can promote the translation from a circRNA expressing another protein (in this case was a circRNA-encoded peptide), demonstrating the versatility of this approach. Moreover, the authors showed that their approach can promote translation in other cell lines.
This is an important and solid study that identified sequences that can improve circRNA translation and that as or more importantly are very short and hence are suitable for generating of efficient protein expressing circRNAs. This manuscript fills an important gap in the field, and it is highly significant. The study is well controlled, the rationale clear and the results conclusive with no major flaws. While this is a minor concern as the vector has been used before, it will greatly improve the quality of the paper if the authors could just verify that the vector only generates circRNA molecules and not linear concatamers. To do so the authors can focus only in their control and the most optimal transcripts and perform northern blot or well controlled RNAseR experiments to show that all RNA molecules containing the back splicing junction are circular.
Minor comments:
While other studies have identified sequences that can drive circRNA translation, this study has done a great job identifying a very short sequence and additional requirements for optimal translation. This is an important study that will be of high interest for the molecular, cell biology and general biology communities.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
A detailed response to the reviewer comments has been uploaded as a separate file. It contains several embedded figures that cannot be shown through this posting option
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Hamadou, Alunno et al. have found evidence for the notion that although translational regulation plays a key role in determining cell behavior, few studies have explored how single nucleotide polymorphisms (SNPs) affect mRNA translation. They developed a method to analyze allele-specific expression in both total and polysome-associated mRNA using RNA-seq data from HCT116 cells. This approach revealed 40 potential "tranSNPs"-SNPs linked to differences in translation between alleles. One SNP, rs1053639 (T/A) in the 3' untranslated region of the DDIT4 gene, was found to influence translation: the T allele was more often associated with polysomes. Cells engineered to carry the TT genotype produced more DDIT4 protein than those with the AA genotype, especially when exposed to stressors like Thapsigargin or Nutlin that boost DDIT4 transcription. The authors found that the RNA-binding protein RBMX mediates this allele-specific protein expression. Knocking down RBMX in TT cells lowered DDIT4 protein levels to those seen in AA cells. Functionally, TT cells suppressed mTORC1 activity more effectively under ER stress, whereas AA cells had a growth advantage in cell culture and in zebrafish models. In human cancer data from TCGA, individuals with the AA genotype had poorer outcomes under a recessive genetic model.
The manuscript needs major revision due to additional data interpretation, lack of statistical analysis, and lack of mechanistic and causal insights. The paper is overall correlative and descriptive and has not enough data to claim a translation regulation aspect of DDIT4 and the protein product to cause the observed genotypic differences stemming from a SNP in the 3' UTR. The paper reads as a collection of individual findings that do not seem to be very cohesive and ranges from polysome-seq, RBP binding, ER stress, mTOR activity, cellular co-culture tumor models and zebrafish tumor models. I wish the authors would have focused on one aspect and described one finding well. Without addressing these fundamental concerns, the study's core claims regarding p53-dependent responses in cancer remain unsubstantiated. Overall, this reviewer supports the publication in a Review Commons journal dependent on that the points of criticism are adequately addressed in the course of a major revision.
Major comments:
Minor comment:
The manuscript needs major revision due to additional data interpretation, lack of statistical analysis, and lack of mechanistic and causal insights. The paper is overall correlative and descriptive and has not enough data to claim a translation regulation aspect of DDIT4 and the protein product to cause the observed genotypic differences stemming from a SNP in the 3' UTR. The paper reads as a collection of individual findings that do not seem to be very cohesive and ranges from polysome-seq, RBP binding, ER stress, mTOR activity, cellular co-culture tumor models and zebrafish tumor models. I wish the authors would have focused on one aspect and described one finding well. Without addressing these fundamental concerns, the study's core claims regarding p53-dependent responses in cancer remain unsubstantiated. Overall, this reviewer supports the publication in a Review Commons journal dependent on that the points of criticism are adequately addressed in the course of a major revision.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
In this manuscript, Hamadou et al. describe the functional characterization of a 3'UTR SNP (rs1053639) in the DDIT4 gene that influences mRNA localization and translation. The authors use polysome profiling, isogenic HCT116 clones, and molecular assays to link the SNP to allele-specific protein expression, proposing a mechanistic role for RBMX and potentially m6A. The manuscript is clearly written and presents compelling evidence to support the authors conclusion.
Major Comments:
Minor Comments:
The authors present a novel and sound pipeline to identify SNPs that regulate mRNA translation using allelic differences in polysome association. Using this approach, they focus on rs1053639 in the 3'UTR of DDIT4 and provide convincing evidence of its impact on mRNA localization and protein expression in HCT116 cells. While the molecular findings are robust, the biological consequences appear relatively modest, and the proposed clinical relevance remains speculative at this stage.
Overall, the study will be of primary interest to a specialized audience of researchers in the fields of post-transcriptional regulation, RNA biology, and functional genomics. The proof-of-concept framework may also attract broader interest for its potential applications in understanding non-coding genetic variation in cancer biology.
Reviewer expertise: p53 biology, molecular cancer biology
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This study investigates the role of a 3'UTR SNP variant in DDIT4 mRNA on allele specific expression at post transcriptional level. The authors have previously developed an experimental approach to identify differences in allele specific transcript distribution in polysomes vs. This was done using polysome profiling combined with RNA-seq analysis of polysome associated and total RNA fractions. This systematic approach identified 40 candidate transcripts exhibiting differential polysome association between reference and variant alleles, indicating post transcriptional effects. Focusing on DDIT4, the study demonstrated that the SNP variant alters subcellular mRNA localization patterns between cytoplasm and nucleus through an impaired interaction with a specific RNA binding protein. Since DDIT4 functions as a negative regulator of mTORC1 signalling, the study examined the mTOR pathway status in homozygous reference and variant genotypes. Using genome-edited cell lines revealed enhanced proliferative capacity of the homozygous AA variant in both co-culture assays and zebrafish xenograft models. I agree with the authors that we don't know much about allele specific effects on mRNA translation mechanisms. However this study doesn't provide much evidence for translational effects either because the differences appear to be mostly due to the impaired export of the variant RNA from the nucleus. Irrespective, the findings are very important as they show how genetic variants in non-coding regions can result in changes of expression at posttranscriptional level.<br /> A comprehensive suite of experimental approaches was utilized to systematically assess both the SNP's impact on mRNA translation and the gene specific functional consequences for DDIT4. The manuscript is well written and presents the work with great clarity.
Major comments
"HCT116, about 11% of genes with analyzable heterozygous SNPs show a difference in AF between paired total and polysome-bound mRNAs, suggesting allele-specific post-transcriptional and translational control." For the remaining candidate transcripts that did not undergo targeted experimental validation like DDIT4, it remains possible that the observed allele specific translational effects could be attributed to other SNPs located elsewhere within these transcripts or to combinatorial effects involving multiple variants. Have the authors considered this possibility? The authors employed RNA probes designed to mimic the secondary structures of the T and A alleles of endogenous DDIT4 mRNA. Could you clarify the exact composition of these probes, do they contain a partial DDIT4 3'UTR sequence? Is it possible that the probes lack critical sequences required for complete protein recognition? Figure 3A - the authors suggest that "in the mock condition, AA cells showed a slight reduction in translation efficiency for the DDIT4 mRNA, as revealed by higher relative abundance in lighter polysomes (fraction 9)" I am not convinced that this is the case, first because the number of ribosomes per mRNA doesn't necessarily reflect translation efficiency and also the TT seems to have increased monosome fraction, and overall to me the profile suggests of slightly reduced translation for TT. Was the nucleotide sequence of the binding site of RBMX determined and if so is this sequence present within the DDIT4 3'UTR?
Minor
Could the authors maybe define what is meant by "analyzable" SNPs or genes? What was the rationale for the selection of HCT116 cells, from a quick search it appears that DDIT4 effects on mTORC1 inhibition could be cell type specific ("mediates mTORC1 inhibition in fibroblasts and thymocytes, but not in hepatocytes"), have the authors considered other cell types Results section 2: Editing of HCT116 cell... I appreciate the clear methodological explanations provided in this section; however, the manuscript might benefit from more concise organization with substantial portions of this descriptive content relocated to the Methods section. Regarding statistical presentation, I recommend reporting exact significance values rather than using threshold indicators (ns, , *, etc.). This approach provides more informative and transparent statistical reporting as differences between "non-significant" and "significant" designations can be minimal neighbouring p-values that fall on opposite sides of arbitrary thresholds and may be misleadingly interpreted. For instance in Figure 2D, the comparison between TT and AA genotypes may approach statistical significance, and displaying the actual p-values would allow readers to better assess the strength of evidence. Fig 3 What is the significance of the control mRNAs? According to the plots it seems as if these also have variants TT/AA? Figure 5A why does AA clone 6 look so different on the gel? "rs1053639 genotype, a relatively common SNP" - what is the estimated frequency of the SNP?
It is a substantial study and a very interesting story. The findings will be of interest for a broad audience, because it combines elements of basic research and clinical significance. The work allows for interpretation of an allele specific genomic variant outside of the coding region and it reveals the importance of similar characterisation of other SNPs.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Response to the Reviews
We thank the reviewers for their input and detailed feedback, which has helped us improve both the manuscript and the Microscopy Nodes software. Based on the comments, we have implemented new features, currently available as version 2.2.1 of Microscopy Nodes. We have edited the text and figures of the manuscript to reflect these changes and add clarification where needed.
Reviewer #1
Evidence, reproducibility and clarity
*The work by Gros et al. presents a paper introducing Microscopy Nodes, a new plugin for Blender 3D visualization software designed to import and visualize multi-dimensional (up to 5D) light and electron microscopy datasets. Given that Blender is not directly suited for such tasks, this plugin significantly simplifies the process, making its visualization engine accessible to a wide range of researchers without prior knowledge of Blender. The plugin supports importing volumes and labels from generic TIF or modern OME-Zarr image formats and includes supplementary video tutorials on YouTube to facilitate basic understanding of the visualization workflows.
Major comments: - The manuscript suggests that Microscopy Nodes can easily handle large datasets, as evidenced by the showcases. However, in my personal tests, I was unable to import a moderate TIF stack of about 5GB, which is considerably smaller than the showcased datasets. Post-import, a data cube was displayed, but the Blender interface became unresponsive. The manuscript should include a section stating limitations and addressing issues and providing suggestions for visualization of large datasets.*
We want to thank the reviewer for this valuable comment, which led us to find a core issue in Blender’s large data handling. Specifically, Blender’s rasterized pipeline causes issues with > 4 GiB of data loaded. This issue does not occur in the raytraced (Cycles) renderer, which is why we had not previously encountered it.
To address this, we have extended the reloading workflow of Microscopy Nodes to provide a workaround for this. If the data is larger than 4 Gibibytes (GiB) (per timepoint, or per timepoint per channel), Microscopy Nodes now automatically downsamples these data during import. While using these downsampled options is recommended for adjusting the visualization settings, the user can then still make their animation and reload their data to the largest scale for the final render by using the raytraced (Cycles) renderer. Additionally, we have raised this bug with the core Blender developers, and hope to work this out in the long term (blender/blender#136263).
We reflect these changes in the manuscript in the segment:
“Blender currently has a notable limitation that its default ‘quick’ rasterized rendering engines (such as ‘EEVEE’, but also the viewport ‘Surface’ and ‘Wireframe’ modes) do not support more than 4 Gibibytes (GiB) of volumetric data. The raytracing render mode ‘Cycles’, however, can handle large volumetric data. To allow users with large data to flexibly use Microscopy Nodes, we implemented a reloading scheme, where one first loads a smaller version of the data (under 4 GiB per timeframe for all loaded channels combined) - and only upon final render in Cycles, exchange it for the full/larger scale copy (Fig 3A). This downscaling of data offers additional benefits as it allows for fast adjustment of the render settings on e.g. a personal computer which can eventually be transferred to a larger workstation or HPC cluster for the final render at full resolution. This feature is critical as working in Cycles with larger files requires sufficient RAM to fit the (temporary) VDB files comfortably. For example, multiple figures in this manuscript were made on a 32GB RAM M1 Macbook Pro (Fig 1A, Video SV1, Fig 1D, Figure 2A-D, Fig S2A-B), but for larger data or long movies the movies were made on workstations or prepared on a laptop and then transferred to an HPC cluster for final rendering.”
* - The feature of importing Zarr-datasets over HTTP is great, but the import process was very slow in my tests, even on a robust network. For reference, loading 1.8 GB of the PRPE1_4x dataset at s1 level took 52 minutes. This raises concerns about potential code issues and general usability of the suggested workflow.*
We believe that this loading time may have been caused by the same issue that plagued all of our datasets of >4GB outside of the raytraced mode, as we have not seen loading issues like that. Moreover, Microscopy Nodes now supports Zarr version to Zarr 3/OME-Zarr 0.5, which allows ‘sharded’ Zarr datasets, which should be even faster at loading large blocks of data at the same time, as Microscopy Nodes does.
- The onsite documentation is a bit outdated and fails to fully describe the plugin settings.
We have updated our documentation to offer new written tutorials, which include full start-up tutorials, but also for some key extra instructions.
- The YouTube tutorials feature an outdated version of the plugin, which could confuse the general microscopy audience. These should be updated to better align with the current plugin functionality. Additionally, using smaller, easily accessible datasets for these tutorials would improve user testing experiences. Hosting complete (downsampled) demo project folder on platforms like zenodo.org could also enhance usability of such tutorials.
We have made a new series of YouTube tutorials that align with the current interface of Microscopy Nodes. These tutorials include public datasets, allowing users to follow along easily. We have chosen to also retain the older tutorials for users running legacy versions of the plugin, as they cover different workflows.
- The manuscript describes a novel dataset used in Fig. 2, but no reference is provided. Additionally, practical implementation of the coloring description for Fig. 2D can be unclear for inexperienced users, necessitating either step-by-step instructions or the provision of downsampled Blender files to aid understanding.
We have now shared the OME-Zarr address in the text (https://uk1s3.embassy.ebi.ac.uk/idr/share/microscopynodes/FIBSEM_dino_masks.zarr), and included this both in the manuscript and the tutorials. Additionally, to guide the implementation and explain the logic behind the coloring we introduced additional panels in Fig S1 and Fig S2 to showcase the shader setups used for this image.
[OPTIONAL] When importing labels, they can be assigned to individual materials only if initially split into multiple color channels. It would be great if the same logic is implemented when those materials are provided as indices within a single color channel. There can be a switch to define the logic used during the import process: e.g. the current one, when the objects are just colored based on a color map, or when they are arranged as individual materials as done when labels are imported from multiple color channels.
We agree with the reviewer and to address this concern with the update to version 2.2, we have implemented a new colorpicking system (See Fig 3B, inset 3, Fig 3C), this allows users to choose between a single color, various continuous, or categorical color maps.
Minor comments: - The manuscript shows nice visualizations of time series, light, and electron microscopy datasets, but in its current state, it is targeted more for light microscopy, where the signal is white. On the other hand, many EM datasets are rendered in inverted contrast (TEM-like), where the signal is black. To render such volume properly, it is needed to go into the Shading tab and flip the color ramp. Would it be possible to perhaps define the data type during import to accommodate various data types or perhaps select the flipped color ramp when the emission mode is switched off? It could make it easier for inexperienced EM users to use the plugin.
To address this, we include new default settings, with ‘invert colormaps on load’ option in the preferences, and default colors per channel (See Fig S4). We have also implemented a new color picking system in version 2.2 (See Fig 3B, inset 3, Fig 3C) that hopefully makes it easier before and after load to change colors.
- It was not completely clear to me whether it is possible to render a single/multiple EM slices using the inverted (TEM-like) contrast. For example, XY, XZ, YZ ortho slices across the volume. The manuscript contains: "This visualization is also supported in Blender, allowing for arbitrary selections of viewing angles (Fig 2B).", but it is not clear how to achieve that.
We introduced an additional explanation in Fig S1A and added a separate density window in the default shader to make this opaque view easier. To get a single slicing plane, users can reduce the scale of the slicing cube in one axis, at it is now also explained in Fig S2B.
- In 3D microscopy, it is quite common to have data with anisotropic voxels. As a result, the surfaces may require smoothing. I was not able to quickly find a way to smooth the surfaces (at least smooth modifiers for surfaces did not work for me). Is it possible to apply smoothing during the import of labels, or alternatively, smoothing of the generated surfaces can be a topic for an additional YouTube video.
The smoothness of the loaded masks can be indirectly affected in the preferences by changing the mesh resolution (changing the relative amount of vertices per pixel), but can be further affected by operations such as the Blender “Smooth” or e.g. the “Smooth by Laplacian” modifiers. To guide the users in doing so, we have included instructions for smoothing in the written tutorials on the website https://aafkegros.github.io/MicroscopyNodes/tutorials/surface_smoothing/ .
- It is also typical to have somewhat custom color maps for materials. It would be great if the plugin remembers the previously used color map for labels.
We have implemented new Preference settings, which include default colors and colormaps per channel, improving customization and reproducibility. This new option is described in Figure S4.
* - The pixel size edit box rounds up the values to 2 digits after the dot. Could it be changed to accommodate 3 or 4 digits as the units are um.*
Blender’s interface truncates the display, but stores higher-precision values internally, and become visible when users click or edit the values. We have added support for alternative pixel units to reduce the impact of the truncation.
- Import is not working when: - Start Blender - Select Data storage: with project - Overwrite files: on, set env: on, chunked: on - Select a file to import - Save Blender file - Pressing the Load button gives an error: "Empty data directory - please save the project first before using With Project saving."
We thank the reviewer for finding this bug which is now fixed in version 2.2.
- I was not able to play the downloaded supplementary video 3 using my VLC media player, while it was working fine in a browser. The video can be opened but looks distorted and heavily zoomed in. It may need to be re-saved from a video editor.
We have recompiled this video.
- References 12 and 16 are URL links instead of proper references to articles.
Thanks for catching this mistake in our bibliography. We have corrected this.
Significance
*This work effectively bridges a gap in the availability of tools for 3D microscopy dataset visualization. While many visualization programs exist, the high-quality ones are often expensive and thus not accessible to all researchers. The integration of Blender with Microscopy Nodes democratizes access to high-quality 3D visualization, enabling researchers to explore datasets and models from multiple perspectives, potentially leading to new discoveries and enhancing the understanding of key study findings. Despite its limitations, my experience with the plugin was engaging and useful. I would like to thank the authors for such useful work!
Limitations: - There remains a steep learning curve associated with using Microscopy Nodes, primarily due to Blender's complexity. More comprehensive tutorials could help mitigate this. - The conversion of imported images to Blender's internal 32-bit format results in a 4x increase in data size for 8-bit datasets. - Managing moderate-sized volumes (5-10 GB) can be challenging without clear strategies for effective handling. - The import of Zarr-datasets over the net is notably slow.
Audience: The plugin is suitable for a broad audience with a basic understanding of 3D visualization concepts, providing a solid foundation for exploring Blender's extensive features and options for optimal visualizations.
Reviewer expertise: Light microscopy, electron microscopy, image segmentation and analysis, software development, no experience with Blender*
Reviewer #2
*Evidence, reproducibility and clarity *
*Summary:
The article introduces Microscopy Nodes, a Blender add-on designed to simplify the loading and visualization of 3D microscopy data. It supports TIF and OME-Zarr images, handling datasets with up to five dimensions. The authors present different visualization modes, including volumetric rendering, isosurfaces, and label masks, demonstrating the application in light and electron microscopy. They provide examples using expansion microscopy, electron microscopy, and real-time imaging, highlighting how the tool enhances scientific communication and interactive visualization.
Comments:
However, some key aspects could be improved to enhance usability and reproducibility:
Example datasets: The images used in the YouTube tutorials were not accessible, making it difficult to reproduce the workflows shown in the figures and tutorials. It would be helpful if the authors provided direct links to the datasets or ensured that the same examples used in the tutorials were readily available for replication.*
We created new and updated tutorials and for all new tutorials, the data is now easily available from an S3 server.
Input file specifications: The article does not clearly detail how input files should be formatted. Many users will pre-visualize images in Fiji to convert their original images to a compatible format. It would be beneficial to specify which formats are supported for hyperstack creation, including details on bit depth, dimension ordering, label formats, and metadata compatibility, if applicable.
We have added new documentation on this on the website and in the manuscript. The addon can take 8, 16, and 32 bit data, and any dimension order (with the letters tzcyx) and pixel size. Dimension order and pixel size can be edited in the GUI. This is reflected in the manuscript in the rewritten section in Design and Implementation:
“It can handle 8bit to 32bit integer and floating point data, although all data types will be resaved into 32bit floating point VDB files, which can cause temporary files to take up more space than the original. Microscopy Nodes loads 2D to 5D files of containing data across time, z, y, x and channels, in arbitrary order (can be remapped in the user interface as well, Fig 3B, inset 2). To focus on relevant data, users can clip the time axis, which can be useful for long videos.”
* Hardware requirements: The article does not discuss RAM or hardware constraints in detail. In testing, attempting to load two images into the same project caused the program to freeze (tested on Mac M1). Specifying hardware requirements and limitations would help users manage expectations when working with large datasets.*
We have since found a limitation in the Blender engine that indeed limits the amount of data loaded (see also comment by Reviewer 1). Currently, rasterized engines are capped at 4 GiB, and only the raytraced engine can handle larger data. As such, the Microscopy Nodes pipeline, where one works with small images until it is time to render a final version, and the data is only exchanged for the final render, is still viable. To make this easier, we now also included optional downscaling for Tif images. This is described in the rewritten section on Design and Implementation:
“Blender currently has a notable limitation that its default ‘quick’ rasterized rendering engines (such as ‘EEVEE’, but also the viewport ‘Surface’ and ‘Wireframe’ modes) do not support more than 4 Gibibytes (GiB) of volumetric data. The raytracing render mode ‘Cycles’, however, can handle large volumetric data. To allow users with large data to flexibly use Microscopy Nodes, we implemented a reloading scheme, where one first loads a smaller version of the data (under 4 GiB per timeframe for all loaded channels combined) - and only upon final render in Cycles, exchange it for the full/larger scale copy (Fig 3A). This downscaling of data offers additional benefits as it allows for fast adjustment of the render settings on e.g. a personal computer which can eventually be transferred to a larger workstation or HPC cluster for the final render at full resolution. This feature is critical as working in Cycles with larger files requires sufficient RAM to fit the (temporary) VDB files comfortably. For example, multiple figures in this manuscript were made on a 32GB RAM M1 Macbook Pro (Fig 1A, Video SV1, Fig 1D, Figure 2A-D, Fig S2A-B), but for larger data or long movies the movies were made on workstations or prepared on a laptop and then transferred to an HPC cluster for final rendering.”
Significance
*General Assessment:
One of the major strengths of this work is its seamless compatibility with Blender, a powerful and widely used animation and 3D rendering tool. Integrating advanced visualization techniques from the animation and graphics industry into scientific imaging opens new possibilities for presenting complex microscopy data in an intuitive and accessible way. Additionally, the support for OME-Zarr is particularly valuable, as this format represents a major shift in bioimaging towards scalable, cloud-compatible, and standardized data storage solutions. The adoption of OME-Zarr facilitates large-scale data handling and improves interoperability across imaging platforms, making this integration a significant step forward for the field. Overall, the greatest strength of the tool lies in its flexibility for rendering microscopy data, but its accessibility for users without Blender experience might be a challenge.
Advance in the Field This work introduces a novel solution to the visualization challenges in microscopy by leveraging Blender's advanced rendering capabilities.
Audience This paper will be of interest to: Bioimage researchers seeking to enhance their microscopy data visualization. Image analysis tool developers interested in integrating advanced visualization into their workflows.
Field of Expertise This review is based on expertise in image analysis, segmentation, and 3D biological data visualization.*
*Reviewer #3 (Evidence, reproducibility and clarity (Required)):
The paper "Microscopy Nodes: Versatile 3D Microscopy Visualization with Blender" presents an easy and accessible approach for microscopists and microscopy users to visualize their data in a different and more controlled way. The authors have developed a plug-in script that enables the integration of complex 3D datasets into Blender, a widely used software for 3D visualization and illustration. By leveraging Blender's advanced rendering engine, the plug-in provides greater control over the scene, enviromint and presentation of the 3D data.
I believe that this development, especially when combined with additional analysis tools can be of a great value for microscopist and advanced users to presenting their 3D data sets.
However, at this stage, the paper does not seem to fully demonstrate the benefits of using Microscopy Nodes. To enhance the paper impact, it would be helpful for the authors to further emphasize and provide examples of how Blender's rendering specifically improves data presentation and, in turn, enhances the understanding of the data compared to existing solutions. Specifically, the authors claim at the end of the introduction that their development provides powerful tools for high-quality, visually compelling presentations, enabling "more effective communication of 3D biological data." I believe this statement should be supported by a figure comparing currently available visualization methods and demonstrating how using Blender enhances data presentation and by which enhances the communication of the results. *
*Additionally, at the end of the first paragraph of the results, the authors say: "These options allow us to combine the data and its analyzed interpretation in the same representation with Microscopy Nodes." However, this capability already exists in currently available software. Aside from now being able to achieve this in Blender, what additional benefits does it offer? *
We now include a new Table 1, to showcases which requirements for visualizing complex biological data are available in different visualization software, and discuss this in the text:
“Although several tools for 3D visualization of bioimages already exist and offer essential features for microscopy data (Table 1), many are proprietary, and open-source alternatives often struggle to deliver a comprehensive user experience, such as advanced animation and annotation controls. Proprietary solutions may offer some of these capabilities, but they are frequently limited by licensing costs, platform restrictions, and a lack of customizability. In contrast, Blender is a mature, well-supported open-source platform with a large community of developers that excels in both animation and visualization. By integrating microscopy-specific functionality through Microscopy Nodes, Blender becomes a uniquely powerful solution that bridges the gap between high-end graphics capabilities and the specialized needs of bioimage visualization.”
Additionally, we attempted to remake Figure 2C and 2D in the EM-field standard software Amira, but were not able to. This is because without an advanced light scattering algorithm, it is very hard to see the depth in the nucleus, and the semi-transparent masks do show each other behind them, but cannot interact with the volume.
We chose not to include this in the actual manuscript, as we are not experts at the Amira software, and will, by the nature of this manuscript, present a challenge that Blender is especially good at, such as here the combination of scattering light and semitransparent masks.
* In the last sentence of the second paragraph of the results, it is stated: "Blender powered by Microscopy Nodes: the ability to combine microscopy data with any 3D illustration in the same 3D environment." Could you please elaborate on the accuracy of the models that can be built and provide guidelines for achieving this using the data coordinates imported by Microscopy Nodes? If the illustrations are purely freehand and do not require specific accuracy, it would be helpful to clarify the advantages of creating them within the same environment rather than separately, as many scientists currently do. Additionally, if the inclusion of 3D model illustrations is one of the key advantages of using Blender, I believe it would be beneficial to present this in a figure rather than only in the supplementary video. *
We thank the reviewer for this comment and agree that in the previously submitted version of Microscopy Nodes, it was very difficult to align objects accurately, as the coordinate space was not transparent. A hurdle in this was the fact that Blender only works well with the unit ‘meters’. To address this issue, we now provide a choice of mapping the physical size to meters, as shown in the new interface (See Fig 3B, inset 5). Here the user can choose from the default ‘px -> cm’ (this will always look fine for a quick look) to options such as ‘nm -> m’ or ‘µm -> m’, which, combined with the new choice for adjusting the object origin upon load, allow users to treat the Blender coordinate space as based on the actual physical scales. Additionally, other Blender addons, such as Molecular Nodes (Reference 25 of the manuscript), also allow for accurate localization for cryo-EM datasets.
We appreciate the note that we should more clearly display the ability to show our illustrations and the data together in the figure and have added a visualization to show this in Figure 1C.
* Reviewer #3 (Significance (Required)):
The significance of the paper at this stage is primarily technical and mainly relevant to the field of microscopy
My field of expertise is microscopy and 3D visualization of models using mainly Maya3D and AMIRA.*
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The paper "Microscopy Nodes: Versatile 3D Microscopy Visualization with Blender" presents an easy and accessible approach for microscopists and microscopy users to visualize their data in a different and more controlled way. The authors have developed a plug-in script that enables the integration of complex 3D datasets into Blender, a widely used software for 3D visualization and illustration. By leveraging Blender's advanced rendering engine, the plug-in provides greater control over the scene, enviromint and presentation of the 3D data.
I believe that this development, especially when combined with additional analysis tools can be of a great value for microscopist and advanced users to presenting their 3D data sets.
However, at this stage, the paper does not seem to fully demonstrate the benefits of using Microscopy Nodes. To enhance the paper impact, it would be helpful for the authors to further emphasize and provide examples of how Blender's rendering specifically improves data presentation and, in turn, enhances the understanding of the data compared to existing solutions.
Specifically, the authors claim at the end of the introduction that their development provides powerful tools for high-quality, visually compelling presentations, enabling "more effective communication of 3D biological data." I believe this statement should be supported by a figure comparing currently available visualization methods and demonstrating how using Blender enhances data presentation and by which enhances the communication of the results.
Additionally, at the end of the first paragraph of the results, the authors say: "These options allow us to combine the data and its analyzed interpretation in the same representation with Microscopy Nodes." However, this capability already exists in currently available software. Aside from now being able to achieve this in Blender, what additional benefits does it offer?
In the last sentence of the second paragraph of the results, it is stated: "Blender powered by Microscopy Nodes: the ability to combine microscopy data with any 3D illustration in the same 3D environment." Could you please elaborate on the accuracy of the models that can be built and provide guidelines for achieving this using the data coordinates imported by Microscopy Nodes? If the illustrations are purely freehand and do not require specific accuracy, it would be helpful to clarify the advantages of creating them within the same environment rather than separately, as many scientists currently do. Additionally, if the inclusion of 3D model illustrations is one of the key advantages of using Blender, I believe it would be beneficial to present this in a figure rather than only in the supplementary video.
The significance of the paper at this stage is primarily technical and mainly relevant to the field of microscopy
My field of expertise is microscopy and 3D visualization of models using mainly Maya3D and AMIRA.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
The article introduces Microscopy Nodes, a Blender add-on designed to simplify the loading and visualization of 3D microscopy data. It supports TIF and OME-Zarr images, handling datasets with up to five dimensions. The authors present different visualization modes, including volumetric rendering, isosurfaces, and label masks, demonstrating the application in light and electron microscopy. They provide examples using expansion microscopy, electron microscopy, and real-time imaging, highlighting how the tool enhances scientific communication and interactive visualization.
Comments:
However, some key aspects could be improved to enhance usability and reproducibility:
Example datasets: The images used in the YouTube tutorials were not accessible, making it difficult to reproduce the workflows shown in the figures and tutorials. It would be helpful if the authors provided direct links to the datasets or ensured that the same examples used in the tutorials were readily available for replication.
Input file specifications: The article does not clearly detail how input files should be formatted. Many users will pre-visualize images in Fiji to convert their original images to a compatible format. It would be beneficial to specify which formats are supported for hyperstack creation, including details on bit depth, dimension ordering, label formats, and metadata compatibility, if applicable.
Hardware requirements: The article does not discuss RAM or hardware constraints in detail. In testing, attempting to load two images into the same project caused the program to freeze (tested on Mac M1). Specifying hardware requirements and limitations would help users manage expectations when working with large datasets.
General Assessment:
One of the major strengths of this work is its seamless compatibility with Blender, a powerful and widely used animation and 3D rendering tool. Integrating advanced visualization techniques from the animation and graphics industry into scientific imaging opens new possibilities for presenting complex microscopy data in an intuitive and accessible way. Additionally, the support for OME-Zarr is particularly valuable, as this format represents a major shift in bioimaging towards scalable, cloud-compatible, and standardized data storage solutions. The adoption of OME-Zarr facilitates large-scale data handling and improves interoperability across imaging platforms, making this integration a a significant step forward for the field. Overall, the greatest strength of the tool lies in its flexibility for rendering microscopy data, but its accessibility for users without Blender experience might be a challenge.
Advance in the Field
This work introduces a novel solution to the visualization challenges in microscopy by leveraging Blender's advanced rendering capabilities.
Audience
This paper will be of interest to: Bioimage researchers seeking to enhance their microscopy data visualization. Image analysis tool developers interested in integrating advanced visualization into their workflows.
Field of Expertise
This review is based on expertise in image analysis, segmentation, and 3D biological data visualization.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The work by Gros et al. presents a paper introducing Microscopy Nodes, a new plugin for Blender 3D visualization software designed to import and visualize multi-dimensional (up to 5D) light and electron microscopy datasets. Given that Blender is not directly suited for such tasks, this plugin significantly simplifies the process, making its visualization engine accessible to a wide range of researchers without prior knowledge of Blender. The plugin supports importing volumes and labels from generic TIF or modern OME-Zarr image formats and includes supplementary video tutorials on YouTube to facilitate basic understanding of the visualization workflows.
Major comments:
[OPTIONAL] When importing labels, they can be assigned to individual materials only if initially split into multiple color channels. It would be great if the same logic is implemented when those materials are provided as indices within a single color channel. There can be a switch to define the logic used during the import process: e.g. the current one, when the objects are just colored based on a color map, or when they are arranged as individual materials as done when labels are imported from multiple color channels.
Minor comments:
The pixel size edit box rounds up the values to 2 digits after the dot. Could it be changed to accommodate 3 or 4 digits as the units are um.
Import is not working when:
This work effectively bridges a gap in the availability of tools for 3D microscopy dataset visualization. While many visualization programs exist, the high-quality ones are often expensive and thus not accessible to all researchers. The integration of Blender with Microscopy Nodes democratizes access to high-quality 3D visualization, enabling researchers to explore datasets and models from multiple perspectives, potentially leading to new discoveries and enhancing the understanding of key study findings. Despite its limitations, my experience with the plugin was engaging and useful. I would like to thank the authors for such useful work!
Limitations:
Audience: The plugin is suitable for a broad audience with a basic understanding of 3D visualization concepts, providing a solid foundation for exploring Blender's extensive features and options for optimal visualizations.
Reviewer expertise: Light microscopy, electron microscopy, image segmentation and analysis, software development, no experience with Blender
Note: This response was posted by the corresponding author to Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Reviewer #1 (Evidence, reproducibility and clarity):
In Arabidopsis, DNA demethylation is catalyzed by a family of DNA glycosylases including DME, ROS1, DML2, and DML3. DME activity in the central cell leads to the hypomethylation of maternal alleles in endosperm. While ROS1, DML2, and DML3 function in vegetative tissues to prevent spreading DNA methylation from TE boundaries, their function in the endosperm was unclear.<br /> Using whole genome methylome analysis, the authors showed that ROS1 prevents hypermethylation of paternal alleles in the endosperm thus promotes epigenetic symmetry between maternal and paternal genomes.<br /> The approach and experimental desighs are appropriate, and the key conclusions are adequately supported by the results.<br /> However, there is not sufficient evidence to support the claim that DME demethylates the maternal allele at ROS1-dependent biallelically-demethylated regions. To clarify the issue, the authors could analyze if there is an overlap between DMRs identified in ros1 endosperm and those identified in dme endosperm using published data. If there is any, the authors could show a genome browser example of DMR including dme data.
Response: Thank you for your insight on our work. To address your concern and further test our model that DME prevents methylation of the maternal allele at regions where ROS1 is prevents methylation of the paternal allele, we turned to the allele-specific bisulfite-sequencing data published in Ibarra et al 2012. These data were from endosperm isolated at 7-8 DAP from aborting seeds of dme-2 +/- (Col-gl) plants pollinated by L_er_. Our analysis of these data is now included in Figures 6 and 7 and Supplemental Figures 13-17. We show that when the loss-of-function allele dme-2 is inherited maternally, average methylation of the maternal allele increases at ROS1-dependent regions (in the revised version of the paper now referred to as ROS1 paternal, DME maternal regions) from less than 10% CG methylation to approximately 40% CG methylation (Fig. 6D), consistent with our previous analysis using the non-allelic Hsieh et al 2009 data (now moved to Supplemental Figure 15). These results thus provide additional evidence that DME removes maternal allele methylation at regions where ROS1 removes paternal allele methylation (compare Fig. 6B and 6D). We included relevant genome browser examples in Figure 7E and Supplemental Figure 14. In the revised version, the relationship between ROS1 and DME is further expanded upon in the text.
Reviewer #1 (Significance):
Endosperm is a tissue unique to flowering plants. Though it is an ephemeral tissue, the endosperm plays essential roles for seed development and germination. The endosperm is also the site genomic imprinting occurs, and it has a distinct epigenomic landscape. This work provides a new insight that ROS1 may antagonize imprinted gene expression in the endosperm. However, it was not shown whether imprinted gene expression is indeed affected in ros1, or whether the ros1 mutation has phenotypic consequences. These results would be useful to discuss the evolution and significance of genomic imprinting.
Response: We agree that the biological significance of ROS1-mediated paternal allele demethylation is presently unknown. We performed RNA-seq on wild-type and ros1 3C and 6C endosperm nuclei, but these data were unfortunately not of high enough quality to include in the manuscript. In the Discussion we suggest that disrupting ROS1-mediated paternal allele demethylation might lead to a gain of imprinting over evolutionary time. In future work we are planning to address potential relationships to gene imprinting using a molecular, RNA-sequencing approach as well as an evolutionary comparative approach. As expected, given the expectation that imprinted genes are associated with a parent-of-origin specific epigenetic mark, we did not find any relationship between known imprinted genes and ROS1-dependent regions that are biallelically-demethylated regions in wild-type endosperm (see lines 362-372).
Reviewer #2 (Evidence, reproducibility and clarity):
SUMMARY
Hemenway and Gehring present evidence that the paternal genome in Arabidopsis endosperm is demethylated at several hundred loci by the DNA glycosylase/lyase ROS1. The evidence is primarily based on analysis of DNA methylation of ros1 mutants and of hybrid crosses where each parental genome can be differentiated by SNPs. I have some comments/questions/concerns, two of them potentially serious, but I think Hemenway and Gehring can address them through additional analyses of data that they already have available and a bit of clarification in writing.
Response: Thank you for your thoughtful review of this study. Your insight and suggestions have helped add clarity to the paper.
MAJOR COMMENTS:
- Could the excess methylation in ros1-3 relative to ros1-7 shown in Figures 1A and 1C be explained by a second mutation in the ros1-3 background that elevates methylation at some loci? Any mutation that increased RdDM at these loci, for example could have this effect. This could confound the identification and interpretation of biallelicly demethylated loci.
Response: We propose a simpler explanation for the additional hypermethylation observed in ros1-3: ros1-3 is a loss-of-function (null) allele whereas ros1-7 is likely a hypomorphic allele. For clarity, we have added a diagram of all of the alleles used in this study as Supplemental Figure 1B. The ros1-3 allele was first described in Penterman et al, PNAS, 2007. It is a T-DNA insertion allele that was isolated in the Ws accession and then backcrossed 6 times to Col-0, greatly minimizing the risk of unlinked secondary mutations being present. There is no genetic evidence that there is another T-DNA insertion in this line. The ros1-7 allele was described in Williams et al, Plos Genet, 2015. It was isolated from the Arabidopsis Col-0 TILLING population and is missense mutation (E956K) in a residue in the glycosylase domain that is conserved among the four DNA glycosylases. It is known that ROS1 transcripts are produced from the ros1-7 allele (Williams et al 2015). We observe less hypermethylation in the ros1-7 background compared to the ros1-3 background, and thus propose that the ros1-7 allele is a hypomorphic allele of ROS1. The use of two independent ros1 mutant alleles for initial endosperm methylation profiling strengthens the findings of our study. Importantly, regions that are hypermethylated in ros1-3 are also hypermethylated in ros1-7, but to a lesser extent, and vice versa (Fig 1D, Supplemental Figs. 3 and 4).
We also use a third allele in this study, ros1-1, which is a nonsense allele in the C24 accession. Notably, we find that the regions are demethylated on both maternal and paternal alleles in wild-type C24 gain DNA methylation primarily on the paternal allele in ros1-1 endosperm (Figure 4C,D and Supplemental Figure 10). This is discussed further in response to your second point.
Given these lines of evidence, a gain-of-function mutation in a methylation pathway, like RdDM, in the ros1-3 background is an unlikely explanation for increased hypermethylation compared to ros1-7. The use of three independent ros1 alleles for methylation profiling, all of which lead to the same conclusions, is a major strength of our study.
- It appears that the main focus of the manuscript, the existence of loci that are paternally demethylated by ROS1, is supported by a set of 274 DMRs. This is a small number relative to the size of the genome and raises suspicions of rare false positives. Even the most stringent p-values that DMR-finding tools report do not guarantee that the DMRs are actually reproducible in an independent experiment. Demonstrating overlap between these 274 DMRs and an independently defined set using a different WT control and different ros1 allele would suffice to remove this concern. It appears that authors already have the needed raw data with ros1-1 and ros1-7 alleles.
Response: First, we should clarify that paternal demethylation by ROS1 is supported by more than the 274 DMRs. All ros1 CG hyperDMRs show an increase in paternal allele methylation in ros1 (Fig. 4B,D). The 274 DMRs are a distinct subset defined as having less methylation on the maternal allele than the paternal allele in ros1 endosperm and where there is no maternal allele hypomethylation in wild-type endosperm (refer to Fig. 5B).
We agree with your sentiments about DMR-finders and we are cautious of relying exclusively on DMR calls when making conclusions. We verify the nature of identified DMRs using metaplots and weighted average comparisons throughout the paper, which we think increases confidence in the conclusions and goes beyond a simple DMR-calling approach.
We argue that we have replicated the major conclusion of the paper, that ROS1 prevents paternal allele hypermethylation at target regions in the endosperm, in the following ways:
Response: Thank you for your feedback and suggestions. We have edited the main text so that only one descriptive name is used for each DMR type throughout the paper. We have also renamed regions for greater clarity. The previous “ROS1-independent, maternally demethylated regions” are now referred to as “DME maternal regions”. The previous “ROS1-_independent, biallelically-demethylated regions” are now referred to as “_ROS1 paternal, DME maternal regions”. These changes provide greater clarity and also emphasize the role of DME at regions that are paternally hypermethylated in ros1. We have added Table 1 to summarize the DMR classes of interest.
MINOR COMMENTS
- The sRNA results in Figure 2B are difficult to interpret because they do not reveal anything about the number of TEs that have siRNAs overlapping them or their flanks. While the magnitude of some of the highest endosperm sRNA peaks is higher than the embryo peaks, that could be explained by a small number of TEs with large numbers of sRNAs. To make this result more interpretable, we also need some information about how many TEs have a significant number of sRNAs associated with them in endosperm and embryo in each region (e.g., middle, 5', 3', and flanks of TEs). What a "significant number of sRNAs" is would be up to the authors to decide based on the distribution of sRNA counts they observe for TEs. Perhaps the top quartile of TEs? Combined with the same analysis done in parallel with non-ROS1 target TEs, this would reveal whether there is any evidence for ROS1 counteracting sRNA-driven methylation spread from TEs.
Response: Thank you for the suggestion. We now present these data and the data for individual TEs underlying the metaplots in Supplemental Figure 7. As suggested by the reviewer, ROS1 TEs do not have uniformly higher levels of sRNA in their flanks in the endosperm compared to the embryo. We have modified our interpretations accordingly.
- The statement "we are likely underestimating the true degree of differential methylation among genotypes" should be validated and partially quantified using a methylation metaplot like Figure 2A, but substitute DMRs for TEs. Related to that, Figure 1B needs an indicator of scale in bp.
Response: We have now included a methylation metaplot over ros1-3 hyperDMRs and ros1-7 hyperDMRs as Supplemental Figure 3 These plots show that indeed there is additional hypermethylation in DMR-proximal regions. We have added a scale bar to Figure 1B and other browser examples in the paper.
- The statement "Over half of ROS1 target regions identified in the ros1-3 mutant endosperm were within 1 kb or intersecting a TE (Fig. 1D)" is hard to interpret without some kind of ROS1 non-target regions or whole-genome control comparison. How different are the numbers in Fig. 1D from a random expectation?
Response: We have now included a control for random regions in Figure 1E. We define these as regions where there was sufficient methylation data coverage and a low enough methylation level in wild-type to detect hypermethylation if it existed.
- The sentence at line 262 is confusing. Is the comparison between dme mutant and ros1 mutant or between different types of regions? And it appears that the comparison value is missing in the "3-5% CG methylation gain..." e.g., "3-5% CG methylation vs 10-20%" or something like that.
Response: This section has been re-written as we now focus on allele-specific dme endosperm methylation data for our comparisons.
- The dme mutant data in Figure 5C appear to be key to the model in Figure 7. The relative impact of the dme mutant in the two types of regions should be quantified.
Response: Thank you for this comment. To further probe our model that DME prevents hypermethylation of the maternal allele at regions where ROS1 is preventing hypermethylation of the paternal allele, we turned to the allele-specific bisulfite-sequencing data published in Ibarra et al 2012 (see also response to reviewer #1). Using these data, we show that when the loss-of-function allele dme-2 is inherited maternally, ROS1 paternal, DME maternal regions (previous referred to as ROS1-_dependent, biallelically-demethylated regions) are CG hypermethylated on the maternal allele (Figure 6D). Thus, these results both replicate the observations made with the Hsieh et al 2009 data, and provide additional evidence that _DME prevents maternal allele hypermethylation at regions were ROS1 is preventing paternal allele hypermethylation. These results have replaced the Hsieh et al 2009 results in Figure 6, and we have moved the analysis of Hsieh et al 2009 data to Supplemental Figure 15.
- Looks like sRNA methods are missing.
Response: Thank you for identifying this. We previously included the reference for the analyzed dataset we used and the method for plotting under an unclear section header. These methods are now in the section “Analysis of average methylation and 24-nt sRNA patterns for features of interest”, and we have added additional reference to the specific dataset we used.
- Supplemental Figure 1 is hard to interpret since it only list gene IDs, not gene names.
Response: As suggested, we have added gene names to this figure.
The last comments are suggestions for increasing the impact of this study:
- Figure 2A and 3B suggest that ROS1 target TEs show demethylation in their flanks but not in the TE themselves. This is an interesting result. If it is true, more DMRs would be expected in the ROS1 target flanks than in the ROS1 target TEs. Reporting how many ROS1 target TEs have DMRs in them and what proportion have DMRs in their flanking 1-Kb regions would answer this question. Given the significance of this result, it also deserves a bit more context: Is the magnitude of increased methylation flanking TEs in ros1 mutant endosperm different than in ros1 mutant leaves or other tissue? Does methylation in TE flanks behave the way in dme mutant endosperm?
Response: We define “ROS1 target TEs” (now referred to more simply as ROS1 TEs) as TEs within 1kb or intersecting a ros1-3 hyperDMR. Consistent with your interpretation, 80% of the TEs in this category do not have a DMR overlapping them, instead they have a TE within 1kb. We now mention this in the text on line 150.
The total level of DNA methylation at ROS1 TEs is lower in the endosperm than in leaf, as DNA methylation levels are overall lower in endosperm than in leaf. The magnitude of increased methylation flanking TEs in ros1 mutant endosperm is not different between the two tissues. This is observable in Supplemental Fig. 5 in the revised version of the paper, and we report this result in the revised text. In the revision we also present methylation profiles of DME TEs in WT and ros1 endosperm (Fig. 7B-D). DME TEs are hypomethylated in both the body and flanks in WT and ros1.
- The idea of biallelic demethylation has been theoretically suggested in maize to explain weak overlap between endosperm DMRs and imprinting (Gent et al 2022). If that were true in Arabidopsis, then ROS1 target, biallelicly demethylated loci would be less likely to have imprinted expression than maternally demethylated loci. This prediction could be tested using available data in Arabidopsis.
Response: Indeed, as you hypothesize, there are no known imprinted genes (Pignatta et al 2014) associated with biallelically-demethylated, ROS1-dependent regions (now referred to as ROS1 paternal, DME maternal regions). Expectedly, there are imprinted genes associated with maternally-demethylated regions (now referred to as DME regions). 23 imprinted genes identified in the Pignatta et al 2014 study are within 1 kb or intersecting a DME region. This is discussed on lines 364-374.
- There is currently no evidence for biological significance of biallelicly demethylated loci. Knowing where they are in the genome might give some hints. A figure like Fig. 1D but specifically showing the biallelicly demethylated DMRs would be valuable.
Response: This is now included in Figure 7A.
- It is hard to make the comparisons between genotypes and parental genomes in Figure 6 and know what they mean. Maybe a different way of displaying the data would help. Or maybe even a different labeling system could make it a little more accessible.
Response: We have revised this figure (now Fig. 8) in the following ways, which we believe address your comments and clarify the main conclusions:
Figure 8C is now a boxplot comparing methylation of the paternal allele of ROS1 paternal, DME maternal regions (previously referred to as biallelically-demethylated, ROS1-dependent regions) across endosperm ROS1 genotypes. This plot shows increased methylation of paternal alleles when the paternal parent is a ros1 mutant, regardless of whether the resultant F1 endosperm is homozygous or heterozygous for ros1 (columns 3, 4, 6).
Figure 8B remains as a scatterplot, where we can observe significant correlation between individual ROS1 paternal, DME maternal regions in homozygous ros1 endosperm and heterozygous ros1/+ endosperm. Note that paternal allele methylation is higher in homozygous ros1 endosperm for most regions.
Reviewer #2 (Significance):
Demethylation of the maternal genome in endosperm has been the subject of much research because it can result in genomic imprinting of gene expression. The enzymes responsible, DNA glycosylases/lyases, also demethylate DNA in other cell types as well, where DNA methylation is not confined to one parental genome (biallelic or biparental as opposed to uniparental demethylation). To the best of my knowledge, the extent or even existence of biallelelic demethylation in endosperm has not been studied until now (except for a superficial look in a bioRxiv preprint, https://www.biorxiv.org/content/10.1101/2024.07.31.606038v1). Hemenway and Gehring have carried out a thoughtful and detailed analysis of the topic in Arabidopsis at least as far as it depends on the DNA glycosylase ROS1.
A limitation is that the study design would miss biallelic demethylation by any of the other three DNA glycosylases in Arabidopsis. A second limitation is that there is no clear biological significance, just some conjecture about evolution. Nonetheless, given the novelty of the topic, biological significance may follow.
The audience for biallelic DNA demethylation in Arabidopsis endosperm is certainly in the "specialized" category, but its relevance to the larger topic of gene regulation in endosperm will attract a larger audience.
Response: With regard to the other demethylases, note that we also profiled methylation in ros1 dml2 dml3 triple mutant endosperm. We did not find evidence for many DMRs that were present in the triple mutant that were not present in the ros1 single mutant. We do not rule out a function for DML2 or DML3 in the endosperm, but this is not observed at the level of bulk endosperm.
The reviewer is correct that we have shown a molecular phenotype (paternal allele hypermethylation) and not a developmental or morphological phenotype. A function that occurs in one parent but not the other is, to us, exciting. Our thoughts about how this finding might relate to imprinting are indeed speculative, but not wildly so.
Reviewer #3 (Evidence, reproducibility and clarity):
DNA demethylases play a key role in DNA methylation patterning during flowering plant reproduction. The demethylase DME, in particular, is critical for proper endosperm development. While the function of DME in endosperm development has been explored, the contributions of the other demethylases in the same family, ROS1, DML2 and DML3 in Arabidopsis, have not yet been investigated. In vegetative tissues, ROS1 prevents hypermethylation of some loci. In this work, Hemenway and Gehring explore whether ROS1, DML2 and DML3 also affect DNA methylation patterns in endosperm. Using EM-seq of sorted endosperm nuclei, they show that loss of ROS1 indeed causes hypermethylation of a number of loci, particularly the flanks of methylated transposons, while loss of DML2 and DML3 has minimal additional effect. By obtaining allele-specific EM-seq data through crosses of Col and C24, the authors show that ros1 endosperm hypermethylation is mostly restricted to the paternal allele. The authors propose that at some sites, ROS1 helps bring down paternal methylation levels to match maternal methylation levels, which are typically reduced in endosperm due to DME activity in the female gametophyte prior to fertilization. In a ros1 mutant with paternal hypermethylation, these sites become differentially methylated on the maternal and paternal alleles, resembling imprinted loci. This work convincingly establishes a function for ROS1 in DNA methylation patterning in endosperm. However, I struggled with the clarity of the writing and reasoning in a few places, and would suggest clarification of a few points and additional analyses below.
Response: Thank you for your thoughtful review of our paper. Your questions and suggestions have been invaluable in revising the work.
I think making a few simple changes to streamline nomenclature would improve readability. For example, in the section starting on line 129, the same set of genomic features are called ROS1 target-proximal TEs, TEs that are near a ROS1 target region, and ROS1 target-associated TE regions. Also for example in line 254 "regions that are maternally-demethylated in wild-type endosperm, and are not dependent on ROS1 for proper demethylation" - are these the same as the "ROS1-independent, maternally-demethylated" regions in Fig. 5a? Given how complex these terms are, being consistent throughout the manuscript really helps the reader.
Response: We edited the text and figures so that only one descriptive name is used for each DMR class or region throughout the paper. Thank you for this feedback; these edits have made the paper much clearer.
Is there any notable effect of ros1 on gene expression in endosperm? Endosperm is a terminal tissue, so maintaining DNA methylation boundaries as ROS1 does in vegetative tissues seems less important. It begs the question of why ROS1 is doing this in endosperm, is it just because it's there, or is there an endosperm-specific function? Exploring effects on imprinting would be particularly interesting (does loss of ROS1 'create' imprinted loci at these newly asymmetrically methylated sites?) but probably beyond the scope of the present work.
Response: We agree, the question of the functional consequence of ROS1 activity in the endosperm is something we are keen to address in future work. We performed RNA-seq on wild-type and ros1 3C and 6C endosperm nuclei, but these data were unfortunately not of high enough quality to include in the manuscript. We are in particular interested in this question you have proposed – if loss of ROS1 can ‘create’ imprinted loci. We are planning to address this both using a molecular, RNA-sequencing approach as well as an evolutionary comparative approach. This is an important and exciting future direction.
Is DME expressed in sperm, or is expression of DME affected in ros1 sperm or endosperm? One other explanation for ros1 hypermethylation occurring primarily on the paternal allele is that, potentially, DME can substitute for ROS1 in the central cell where DME is already very active, but not in sperm cells. Related, how well expressed is ROS1 vs. DME in sperm cells?
Response: This is an important series of questions, and something we are very interested in as well. Studies of Arabidopsis pollen have shown that both ROS1 and DME, while they prevent some hypermethylation in sperm, are more active in the vegetative nucleus of pollen than in sperm. ROS1 is expressed at a low level in the microspore and bicellular pollen and DME is expressed at a low level throughout pollen development. We have included Supplemental Fig. 17 with available expression data to make this point in the paper. Likely, any effects of loss of ROS1 or DME on sperm DNA methylation are inherited from precursor cells (Ibarra et al 2012, Calarco et al 2012, Khouider et al 2021). Your proposal that perhaps DME can sub in for ROS1 in the central cell but not in sperm is intriguing. Unfortunately there’s not enough data in the central cell to convincingly address this at this time.
To investigate the relationship between DME and ROS1 in the male germline, we used the bisulfite-sequencing data generated in sperm cells in Khouider et al 2021. We calculated average DNA methylation levels in dme/+, ros1, dme/+;ros1, and wild-type Col-0 sperm cells at ROS1 paternal, DME maternal regions, shown in Supplemental Fig. 18A. We observed little increase in mCG methylation in dme/+ sperm relative to wild-type Col-0 sperm. This is consistent with your proposed model that DME is unable to demethylate these regions outside of the female germline. As expected, there is increased mCG in ROS1 paternal, DME maternal regions in ros1-3 mutant sperm relative to wild-type Col-0 sperm. DME maternal regions are highly methylated in wild-type Col-0 sperm.
Fig 2b shows that ROS1 target-associated TEs are enriched for sRNAs in endosperm relative to embryo, whereas the reverse is true for non-ROS1-assoc TEs. Since TEs are not always well annotated and some may be missing from this analysis, what about trying the reverse analysis - are regions enriched for 24nt sRNAs in endosperm significantly hypermethylated in ros1 endosperm? All regions or only some?
Response: We performed an analysis to address your inquiry and observed a low magnitude increase in DNA methylation in ros1 mutant endosperm at regions defined by Erdmann et al as more sRNA producing in the endosperm relative to the embryo (endosperm DSRs). Endosperm DSRs are generally lowly methylated in wild-type endosperm, as was observed originally in Erdmann et al 2017. Small increases in DNA methylation are observed at endosperm DSRs in all sequence contexts in ros1 endosperm. Overall, this is consistent with ROS1 targets being a subset of sRNA-producing regions in the endosperm. This analysis is now included in Supplemental Fig. 7C.
What is the relationship between previously-defined DME targets and ROS1 targets identified in this paper? DME tends to target small euchromatic TE bodies, whereas Fig. 3 suggests that ROS1 helps prevent methylation spreading on the outer edges of the TEs, rather than in the TE body. Do all DME targets tend to be adjacent to or flanked by ROS1 target sites? Or are the TEs affected by DME (in body) and by ROS1 (at edges) largely nonoverlapping? Fig. 5a suggests that the ROS1-dependent, biallelically-demethylated sites are both DME and ROS1 targets, but how often do these really appear to overlap? More than by chance?
Response: We have sought to address your comments through a series of analyses that we have included in Fig. 7 and Supplemental Fig. 16. We found that ROS1 paternal, DME maternal regions (formerly referred to as ROS1-dependent, biallelically-demethylated regions) and DME maternal regions (formerly referred to as ROS1-independent, maternally-demethylated regions) do not occupy the same genomic regions. However, we do observe some evidence for ROS1 activity in flanking regions of DME targets (Fig. 6A, Fig. 7B-D). To look at TEs specifically, as you suggest, we first identified TEs that were within 1kb or intersecting a DME maternal region. Based on our characterization of these regions, we assume these to be DME-targeted TEs. We then performed ends analysis to see if there was evidence of ROS1 activity at the ends of these TEs. Indeed, at a global level there is a slight hypermethylation of the paternal allele in a ros1 mutant at the end of these DME TEs (Fig. 7B). To better visualize how many DME TEs are showing ROS1 activity at their ends, we then plotted the difference between the median ros1-3 methylation and median Col-0 values in the non-allelic endosperm for each TE in a clustered heatmap (Fig. 7C). The parent-of-origin data does not have enough coverage for clustering in this way, so we used the non-allelic data. A small fraction of “DME TEs” gain methylation in the ros1 mutant endosperm relative to wild-type (Fig. 7C-D).
Are the TEs whose boundaries are demethylated by ROS1 more likely to be expressed in vegetative or endosperm tissues than TEs not affected by loss of ROS1? Expressed TEs likely produce more sRNAs, which would increase RdDM in a way that might need to be more actively countered by ROS1 than transcriptionally silent or evolutionarily older TEs.
Response: This is an interesting line of inquiry, although perhaps out of the scope of our present study. It has been shown that TEs demethylated by ROS1 are targeted by the RdDM pathway in Arabidopsis vegetative tissue (Tang et al 2016). Using data from Erdmann et al 2017, we looked at 24 nt sRNAs at ROS1-TEs in the endosperm and embryo (Supplemental Fig. 7). sRNA production at ROS1 TE-flanking regions is observed in both embryo and endosperm, but clearly not all ROS1 TEs produce 24 nt sRNA production in the seed. Future work comparing sRNA profiles in a ros1 mutant to those of wild-type could inform our understanding of TE spreading in a ros1 mutant, as would a comprehensive analysis of TE expression, again in both a ros1 mutant and in wild-type. It’s unclear to us if the endosperm would be the most informative or useful tissue to perform such analyses in.
Fig6 - as noted in the text, one way to test whether demethylation by ROS1 occurs before or after fertilization is to provide functional ROS1 through only one parent via reciprocal WT x ros-1 crosses, so that the endosperm always has ROS1 but either sperm or central cell does not, and see if this can rescue the paternal hypermethylation. If ROS1 acts prior to fertilization, then paternal ROS1 will rescue ros1 hypermethylation, but maternal ROS1 won't. If after fertilization, then either maternally or paternally supplied ROS1 will rescue the hypermethylation phenotype (assuming both are well expressed). Thus, to distinguish the two, it is sufficient to test whether maternally supplied ROS1 in an otherwise mutant background can rescue the hypermethylation phenotype, which is what is shown in Fig. 6. However, I think it's also important to show that paternally supplied ROS1 can also rescue the hypermethylation phenotype, which is not currently shown. The plots showing no effect on maternal mCG aren't as informative, since maternal methylation levels are mostly unaffected by ros1 anyway. Instead of comparing pairs of samples in a scatterplot, it might be clearer to show paternal mCG across all four comparisons (WT x WT, WT x ros1, ros1 x WT, and ros1 x ros1) side by side in a heatmap, using clustering to group similar behavior.
Response: We have revised this figure, now Fig. 8, in the following ways, which we believe addresses your comments and clarify the main conclusions (see same response to reviewer 2 for point 14):
Figure 8B remains as a scatterplot, where we observe significant correlation between individual ROS1 paternal, DME maternal regions in homozygous ros1 endosperm and heterozygous ros1/+ endosperm. Note that paternal allele methylation is higher in homozygous ros1 endosperm for most regions.
Figure 8C is now a boxplot comparing methylation of the paternal allele of ROS1 paternal, DME maternal regions (previously referred to as biallelically-demethylated, ROS1-dependent regions) across endosperm ROS1 genotypes. This plot shows increased methylation of paternal alleles when the paternal parent is a ros1 mutant, regardless of whether the resultant F1 endosperm is homozygous or heterozygous for ros1 (columns 3, 4, 6).
I would also suggest including a little more information in the main plots rather than only in the figure legends. For example, in Fig 2 including a label of 'ROS1-associated TE' for the two plots on the left, and 'TEs not associated with ROS1' on the right. Or for example in Fig. 3a indicating 'ros1-3 CG hyperDMRs' somewhere on the plot. This would just help make the figures easier to read at a glance. Please add common gene names to figures, instead just the ATG gene ID (Fig. S1a).
Response: Thank you for this feedback, we have made the suggested edits and additional edits of a similar nature.
Minor:<br /> - Fig. 1E is referenced in the text before Fig. 1D<br /> - Fig. S4 and S5 - there are more lines in the plot than the 6 genotypes listed in the legend, do these represent different replicates? If so that should be noted in the legend<br /> - Fig. 1B has no color legend for the different methylation sequence contexts (looks like same as 1A,C but should indicate either in plot or legend)<br /> - Line 42 should be "correspond to TE ends"<br /> - Line 93 "Based on previous studies..." should have references to those studies<br /> - When referring to the protein (rather than the genetic locus or mutant), ROS1 should not be italicized - for example line 130<br /> - Line 150 "we conclude that the loss"<br /> - Should add a y=x line to scatterplots, like those in Fig. 6<br /> - In fig. 1d, it's hard to evaluate the significance of the overlap of ROS1 targets with genes and TEs. Comparing these numbers to a control where the ROS1 targets have been randomly shuffled would help.
Response: We have made edits and additions where requested.
Reviewer #3 (Significance):
In this work, Hemenway and Gehring explore whether ROS1, DML2 and DML3 also affect DNA methylation patterns in endosperm. Using EM-seq of sorted endosperm nuclei, they show that loss of ROS1 indeed causes hypermethylation of a number of loci, particularly the flanks of methylated transposons, while loss of DML2 and DML3 has minimal additional effect. By obtaining allele-specific EM-seq data through crosses of Col and C24, the authors show that ros1 endosperm hypermethylation is mostly restricted to the paternal allele. The authors propose that at some sites, ROS1 helps bring down paternal methylation levels to match maternal methylation levels, which are typically reduced in endosperm due to DME activity in the female gametophyte prior to fertilization. In a ros1 mutant with paternal hypermethylation, these sites become differentially methylated on the maternal and paternal alleles, resembling imprinted loci. This work convincingly establishes a function for ROS1 in DNA methylation patterning in endosperm. However, I struggled with the clarity of the writing and reasoning in a few places, and would suggest clarification of a few points and additional analyses.
Response: Thank you for your comments. We have worked on streamlining the text and analysis.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
DNA demethylases play a key role in DNA methylation patterning during flowering plant reproduction. The demethylase DME, in particular, is critical for proper endosperm development. While the function of DME in endosperm development has been explored, the contributions of the other demethylases in the same family, ROS1, DML2 and DML3 in Arabidopsis, have not yet been investigated. In vegetative tissues, ROS1 prevents hypermethylation of some loci. In this work, Hemenway and Gehring explore whether ROS1, DML2 and DML3 also affect DNA methylation patterns in endosperm. Using EM-seq of sorted endosperm nuclei, they show that loss of ROS1 indeed causes hypermethylation of a number of loci, particularly the flanks of methylated transposons, while loss of DML2 and DML3 has minimal additional effect. By obtaining allele-specific EM-seq data through crosses of Col and C24, the authors show that ros1 endosperm hypermethylation is mostly restricted to the paternal allele. The authors propose that at some sites, ROS1 helps bring down paternal methylation levels to match maternal methylation levels, which are typically reduced in endosperm due to DME activity in the female gametophyte prior to fertilization. In a ros1 mutant with paternal hypermethylation, these sites become differentially methylated on the maternal and paternal alleles, resembling imprinted loci. This work convincingly establishes a function for ROS1 in DNA methylation patterning in endosperm. However, I struggled with the clarity of the writing and reasoning in a few places, and would suggest clarification of a few points and additional analyses below.
I think making a few simple changes to streamline nomenclature would improve readability. For example, in the section starting on line 129, the same set of genomic features are called ROS1 target-proximal TEs, TEs that are near a ROS1 target region, and ROS1 target-associated TE regions. Also for example in line 254 "regions that are maternally-demethylated in wild-type endosperm, and are not dependent on ROS1 for proper demethylation" - are these the same as the "ROS1-independent, maternally-demethylated" regions in Fig. 5a? Given how complex these terms are, being consistent throughout the manuscript really helps the reader.
Is there any notable effect of ros1 on gene expression in endosperm? Endosperm is a terminal tissue, so maintaining DNA methylation boundaries as ROS1 does in vegetative tissues seems less important. It begs the question of why ROS1 is doing this in endosperm, is it just because it's there, or is there an endosperm-specific function? Exploring effects on imprinting would be particularly interesting (does loss of ROS1 'create' imprinted loci at these newly asymmetrically methylated sites?) but probably beyond the scope of the present work.
Is DME expressed in sperm, or is expression of DME affected in ros1 sperm or endosperm? One other explanation for ros1 hypermethylation occurring primarily on the paternal allele is that, potentially, DME can substitute for ROS1 in the central cell where DME is already very active, but not in sperm cells. Related, how well expressed is ROS1 vs. DME in sperm cells?
Fig 2b shows that ROS1 target-associated TEs are enriched for sRNAs in endosperm relative to embryo, whereas the reverse is true for non-ROS1-assoc TEs. Since TEs are not always well annotated and some may be missing from this analysis, what about trying the reverse analysis - are regions enriched for 24nt sRNAs in endosperm significantly hypermethylated in ros1 endosperm? All regions or only some?
What is the relationship between previously-defined DME targets and ROS1 targets identified in this paper? DME tends to target small euchromatic TE bodies, whereas Fig. 3 suggests that ROS1 helps prevent methylation spreading on the outer edges of the TEs, rather than in the TE body. Do all DME targets tend to be adjacent to or flanked by ROS1 target sites? Or are the TEs affected by DME (in body) and by ROS1 (at edges) largely nonoverlapping? Fig. 5a suggests that the ROS1-dependent, biallelically-demethylated sites are both DME and ROS1 targets, but how often do these really appear to overlap? More than by chance?
Are the TEs whose boundaries are demethylated by ROS1 more likely to be expressed in vegetative or endosperm tissues than TEs not affected by loss of ROS1? Expressed TEs likely produce more sRNAs, which would increase RdDM in a way that might need to be more actively countered by ROS1 than transcriptionally silent or evolutionarily older TEs.
Fig6 - as noted in the text, one way to test whether demethylation by ROS1 occurs before or after fertilization is to provide functional ROS1 through only one parent via reciprocal WT x ros-1 crosses, so that the endosperm always has ROS1 but either sperm or central cell does not, and see if this can rescue the paternal hypermethylation. If ROS1 acts prior to fertilization, then paternal ROS1 will rescue ros1 hypermethylation, but maternal ROS1 won't. If after fertilization, then either maternally or paternally supplied ROS1 will rescue the hypermethylation phenotype (assuming both are well expressed). Thus, to distinguish the two, it is sufficient to test whether maternally supplied ROS1 in an otherwise mutant background can rescue the hypermethylation phenotype, which is what is shown in Fig. 6. However, I think it's also important to show that paternally supplied ROS1 can also rescue the hypermethylation phenotype, which is not currently shown. The plots showing no effect on maternal mCG aren't as informative, since maternal methylation levels are mostly unaffected by ros1 anyway. Instead of comparing pairs of samples in a scatterplot, it might be clearer to show paternal mCG across all four comparisons (WT x WT, WT x ros1, ros1 x WT, and ros1 x ros1) side by side in a heatmap, using clustering to group similar behavior.
I would also suggest including a little more information in the main plots rather than only in the figure legends. For example, in Fig 2 including a label of 'ROS1-associated TE' for the two plots on the left, and 'TEs not associated with ROS1' on the right. Or for example in Fig. 3a indicating 'ros1-3 CG hyperDMRs' somewhere on the plot. This would just help make the figures easier to read at a glance. Please add common gene names to figures, instead just the ATG gene ID (Fig. S1a).
Minor:
In this work, Hemenway and Gehring explore whether ROS1, DML2 and DML3 also affect DNA methylation patterns in endosperm. Using EM-seq of sorted endosperm nuclei, they show that loss of ROS1 indeed causes hypermethylation of a number of loci, particularly the flanks of methylated transposons, while loss of DML2 and DML3 has minimal additional effect. By obtaining allele-specific EM-seq data through crosses of Col and C24, the authors show that ros1 endosperm hypermethylation is mostly restricted to the paternal allele. The authors propose that at some sites, ROS1 helps bring down paternal methylation levels to match maternal methylation levels, which are typically reduced in endosperm due to DME activity in the female gametophyte prior to fertilization. In a ros1 mutant with paternal hypermethylation, these sites become differentially methylated on the maternal and paternal alleles, resembling imprinted loci. This work convincingly establishes a function for ROS1 in DNA methylation patterning in endosperm. However, I struggled with the clarity of the writing and reasoning in a few places, and would suggest clarification of a few points and additional analyses
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary
Hemenway and Gehring present evidence that the paternal genome in Arabidopsis endosperm is demethylated at several hundred loci by the DNA glycosylase/lyase ROS1. The evidence is primarily based on analysis of DNA methylation of ros1 mutants and of hybrid crosses where each parental genome can be differentiated by SNPs. I have some comments/questions/concerns, two of them potentially serious, but I think Hemenway and Gehring can address them through additional analyses of data that they already have available and a bit of clarification in writing.
Major comments:
Minor comments
The last comments are suggestions for increasing the impact of this study:<br /> 11. Figure 2A and 3B suggest that ROS1 target TEs show demethylation in their flanks but not in the TE themselves. This is an interesting result. If it is true, more DMRs would be expected in the ROS1 target flanks than in the ROS1 target TEs. Reporting how many ROS1 target TEs have DMRs in them and what proportion have DMRs in their flanking 1-Kb regions would answer this question. Given the significance of this result, it also deserves a bit more context: Is the magnitude of increased methylation flanking TEs in ros1 mutant endosperm different than in ros1 mutant leaves or other tissue? Does methylation in TE flanks behave the way in dme mutant endosperm?<br /> 12. The idea of biallelic demethylation has been theoretically suggested in maize to explain weak overlap between endosperm DMRs and imprinting (Gent et al 2022). If that were true in Arabidopsis, then ROS1 target, biallelicly demethylated loci would be less likely to have imprinted expression than maternally demethylated loci. This prediction could be tested using available data in Arabidopsis.<br /> 13. There is currently no evidence for biological significance of biallelicly demethylated loci. Knowing where they are in the genome might give some hints. A figure like Fig. 1D but specifically showing the biallelicly demethylated DMRs would be valuable.<br /> 14. It is hard to make the comparisons between genotypes and parental genomes in Figure 6 and know what they mean. Maybe a different way of displaying the data would help. Or maybe even a different labeling system could make it a little more accessible.
Demethylation of the maternal genome in endosperm has been the subject of much research because it can result in genomic imprinting of gene expression. The enzymes responsible, DNA glycosylases/lyases, also demethylate DNA in other cell types as well, where DNA methylation is not confined to one parental genome (biallelic or biparental as opposed to uniparental demethylation). To the best of my knowledge, the extent or even existence of biallelelic demethylation in endosperm has not been studied until now (except for a superficial look in a bioRxiv preprint, https://www.biorxiv.org/content/10.1101/2024.07.31.606038v1). Hemenway and Gehring have carried out a thoughtful and detailed analysis of the topic in Arabidopsis at least as far as it depends on the DNA glycosylase ROS1.
A limitation is that the study design would miss biallelic demethylation by any of the other three DNA glycosylases in Arabidopsis. A second limitation is that there is no clear biological significance, just some conjecture about evolution. Nonetheless, given the novelty of the topic, biological significance may follow.
The audience for biallelic DNA demethylation in Arabidopsis endosperm is certainly in the "specialized" category, but its relevance to the larger topic of gene regulation in endosperm will attract a larger audience.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In Arabidopsis, DNA demethylation is catalyzed by a family of DNA glycosylases including DME, ROS1, DML2, and DML3. DME activity in the central cell leads to the hypomethylation of maternal alleles in endosperm. While ROS1, DML2, and DML3 function in vegetative tissues to prevent spreading DNA methylation from TE boundaries, their function in the endosperm was unclear.
Using whole genome methylome analysis, the authors showed that ROS1 prevents hypermethylation of paternal alleles in the endosperm thus promotes epigenetic symmetry between maternal and paternal genomes.<br /> The approach and experimental desighs are appropriate, and the key conclusions are adequately supported by the results.
However, there is not sufficient evidence to support the claim that DME demethylates the maternal allele at ROS1-dependent biallelically-demethylated regions. To clarify the issue, the authors could analyze if there is an overlap between DMRs identified in ros1 endosperm and those identified in dme endosperm using published data. If there is any, the authors could show a genome browser example of DMR including dme data.
Endosperm is a tissue unique to flowering plants. Though it is an ephemeral tissue, the endosperm plays essential roles for seed development and germination. The endosperm is also the site genomic imprinting occurs, and it has a distinct epigenomic landscape. This work provides a new insight that ROS1 may antagonize imprinted gene expression in the endosperm. However, it was not shown whether imprinted gene expression is indeed affected in ros1, or whether the ros1 mutation has phenotypic consequences. These results would be useful to discuss the evolution and significance of genomic imprinting.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer comment: *“The authors did not clarify whether the observed protection to PTZ-induced convulsions after mild TBI is due to the reduced size of gap junctions and/or increased activity in hemichannels.” And “The super-resolution imaging only assesses Cx43 gap junction plaque size and density but not the non-junctional portion of Cx43.” *
Response and planned revision: To determine whether seizure protection in Cx43 S368A mice is due to reduced gap junction plaque density or reduced hemichannel function, we will conduct solubility assays to assess the ratio of insoluble (junctional) to soluble (cytoplasmic/hemichannel) Cx43 in Cx43S368A and C57BL/6 control mice after TBI/sham (as in Fig. 2A-D currently only in C57BL/6 control mice). In parallel, we will perform EtBr uptake assays in acute brain slices from Cx43S368A and C57BL/6 control animals to assess hemichannel function.
Additionally, we will include super-resolution images without background subtraction, which show diffuse staining indicative of soluble Cx43. Of note, even at super-resolution individual gap junctions or hemichannels cannot be resolved. They appear as diffuse signal (currently not visible in our super-resolution images due to image deconvolution and background substration performed to isolate Cx43 plaques). Super-resolution imaging was used to count Cx43 gap junction plaque densities and size. Cx43 gap junction plaques are dense accruals of Cx43 immunostaining reminiscent functional and closed gap junctions. Complimentary experiments measured soluble (cytoplasmic Cx43 and hemichannels) and insoluble Cx43 (gap junctions) using biochemistry (Fig. 2A-D).
Reviewer comment: “The immunofluorescent images for Fig. 2E and Fig. 5 were not counterstained for astrocytes or cell membrane. How can the authors be sure that these are expressed by astrocytes and not other cells in the brain?”
Response and planned revision: Cx43 is predominantly expressed in astrocytes, with expression levels 10–100 times higher than in brain endothelial cells (e.g., Zhang et al., 2014; Vanlandewijck et al., Nature, 2018). As shown in Supplementary Fig. 2, our immunohistochemistry data reveal no overlap between Cx43 and endothelial cell markers, confirming that our staining protocol does not detect Cx43 in endothelial cells. Instead, the apparent localization of Cx43 along blood vessels reflects expression in astrocytic endfeet, which closely ensheath the vasculature. To further support this conclusion, we will conduct quantitative co-localization analyses of Cx43 with markers for neurons, microglia, oligodendrocytes, and NG2 glia in both Cx43S368A and C57BL/6 control mice. Additionally, we will include plots generated from publicly available single-cell RNA sequencing datasets to show that Cx43 mRNA is highly enriched in astrocytes and present at much lower levels in endothelial cells of the brain vasculature.
Reviewer comment about developmental contributions to the phenotype of Cx43 S368A animals.
Response: We cannot exclude a potential developmental component to the observed seizure protection in Cx43S368A mice. We included discussion of this possibility in the revised manuscript.
Reviewer comments indicative of a lack of clarity around rationale and intent of specific experiments.
Response: We thoroughly revised the Results section to explicitly state the rationale and purpose of each experiment. For example:
Reviewer comment: “The immunofluorescent images for Fig. 1D and E were taken at low resolution compared to the Cx43 puncta size. This does not allow accurate quantification of the Cx43 GJs or HCs.”
Response: The purpose of this experiment was to assess the heterogeneity of Cx43 expression (both junctional and non-junctional portions) with spatial resolution across a larger brain area. Complementary experiments here are quantification of protein amounts using western blot (Fig. 1B), quantification of junctional versus non-junctional Cx43 using the solubility assay and quantification of Cx43 plaques using super-resolution imaging (Fig. 2).
Reviewer comment: “TBI did not change Cx43 plaque size or density (Fig. 5). What was the rationale for examining the effects in the S368A mutant?”
Response: We found an increase in phosphorylated Cx43 at ____S____368 after TBI and Cx43__S368A mutants are protected from seizures after administration of PTZ suggesting an important role for this specific Cx43 phosphorylation site in pathology. __We discussed in the manuscript that “in cardiovascular infection/disease has demonstrated maintenance of gap junction coupling (Gy et al., 2011; Padget et al., 2024) while reduced hemichannel opening probability was reported (Hirschhäuser et al., 2021) in Cx43S368A mice”, suggesting that the protective phenotype is likely due to modification of either Cx43 gap junctions or hemichannels. However, functional consequences on Cx43 biology upon phosphorylation at S368 or lack thereof in the Cx43S368A mutant remain unexplored in the brain. Cx43 plaque size and density are reflective of Cx43 gap junctions and was therefore examined in Cx43S368A mice to reveal potential mechanism by which this mouse mutant is protected from seizures (even in the absence of TBI).
Reviewer comment: * “The IC50 for Tat-Gap19 for Cx43 HC is ~7 μM (Tocris). How can using it at 2 μM be effective?”*
Response: We reviewed our lab records and confirmed that 2 μM was a typographical error. The actual concentration used was 200 μM. This is consistent with the dose-response literature for astrocytes (e.g., Walrave et al., Glia 2018; Abudara et al., Front. Cell. Neurosci. 2014). We now included these references in the manuscript.
Reviewer comment: “Unclear whether mice in Fig. 4C received TBI.”
Response: We clarified that these mice were naïve, i.e. not subjected to TBI or sham procedures. This is now explicitly stated in both the Methods and the Results.
Reviewer comment: “CBX or Tat-Gap19 do not affect the phosphorylation state of Cx43.”
Response: We clarified that we used CBX and Tat-Gap19 as established gap junction and hemichannel blockers, irrespective of phosphorylation state. We now noted that Tat-GAP19 is a Cx43 mimetic peptide to specifically block Cx43 hemichannels.
Reviewer comment: “It is unclear whether the EtBr quantification in Fig. 3D is for S100β+ astrocytes.”
Response: We clarified that the quantification in Fig. 3D was performed exclusively in S100β+ astrocytes. Although neurons may take up EtBr under inflammatory conditions, they do not express Cx43 (as will be shown in Fig. 1 and Supplementary Data).
Reviewer comment: “I believe that the 'W.' in ref 'W. Chen et al., 2018' is unnecessary.”
Response: We will use the journal citation style implemented by a reference manager in the final version of the manuscript.
Reviewer request to include two references related to phosphorylation and hemichannel permeability and the role of gap junctional coupling in epilepsy.
Response: The PNAS reference was added to the manuscript.
That reduction in gap junctional communication is a relevant factor in epilepsy is discussed in the introduction where we also cite original literature of the authors of the proposed review article: “Many pathologies (Gajardo-Gómez et al., 2017; Masaki, 2015; Orellana et al., 2011; Sarrouilhe et al., 2017; Vis et al., 1998; Wang et al., 2018), including traumatic brain injury (TBI) (B. Chen et al., 2017; W. Chen et al., 2019; Wu et al., 2013; Xia et al., 2024) and acquired epilepsy (Bedner et al., 2015; Deshpande et al., 2017; Walrave et al., 2018) present with altered Cx43 regulation, and are often equated with GJ dysfunction.”
We feel that citing the original manuscripts more accurately reflect the current knowledge around the role of Cx43 in the context of epilepsy and other pathologies. Reader’s access to the original literature also highlights the gaps in knowledge more precisely that this manuscript seeks to close.
Reviewer comment: “I think the data of this manuscript is missing a control animal that would present all the compensation changes that occur during development that occur in mice carrying the mutated Cx43. Alternatively, a doable experiment would be the use of inducible KO/KI.”
Response: Previous studies investigating the role of Cx43 in neuronal excitability have primarily used full or conditional knockout models, as described in our introduction. Interestingly, these studies report that global deletion of Cx43 increases seizure susceptibility. However, such models eliminate all Cx43-dependent functions—both junctional and non-junctional—making it difficult to pinpoint the specific mechanisms underlying the observed effects. They do not distinguish whether increased excitability results from loss of gap junction coupling, disruption of hemichannel function, or depletion of cytoplasmic Cx43 signaling. In contrast, our current study does not aim to eliminate Cx43, but instead employs a targeted approach to interrogate the functional significance of a regulatory phosphorylation site, S368. This site is dynamically phosphorylated following TBI and has been previously associated—albeit only through correlative data—with seizure activity and other neuropathologies. By isolating the contribution of this post-translational modification while preserving overall Cx43 expression, our study provides novel mechanistic insight into how phosphorylation modulates Cx43 function and astrocyte-mediated regulation of brain excitability.
We appreciate the thoughtful suggestion to generate a conditional knock-in model to isolate developmental from acute effects of the Cx43 S368A mutation. However, the GJA1 gene locus is not amenable to this type of targeting (we explored this possibility with a . We also considered AAV-mediated CRISPR/dCas9 editing as an alternative, but current limitations in CNS transduction efficiency, promoter specificity, and guide RNA availability for precise point mutation insertion make this approach similarly unfeasible at this stage. Thus, while we acknowledge the developmental caveat (which we now discuss in the manuscript), the current manuscript provides novel and meaningful insight into the role of the Cx43S368 regulatory phosphorylation site in the context of astrocyte biology and seizure susceptibility and forms a strong foundation for future studies.
Thank you again for the opportunity to revise and strengthen our manuscript. We believe these planned experiments and clarifications address the reviewers' concerns in a thorough and scientifically rigorous manner.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript describes interesting findings on the effect of a Cx43 mutant that is not phosphorylated in Ser368. The authors did not clarify whether the observed protection to PTZ-induced convulsions after mild TBI is due to the reduced size of gap junctions and/or increased activity in hemichannels. A limitation of this work is that Cx43 S368A forms smaller gap junctions revealing an important phenotype change and therefore there is no appropriate control unless they generate a cell-specific inducible Cx43 KO.<br /> In previous studies, it has been proposed that reduction in gap junctional communication is a relevant factor in epilepsy, which is not discussed (Please see doi: 10.3390/cells12121669) in the present manuscript. Also, Bao and collaborators have demonstrated that Cx43 hemichannels phosphorlated by PKC present reduced permeability to molecules but continuos permeable to smaller molecules (doi.org/10.1073/pnas.060315410). This is an important finding that should be mentioned in the intruduuction and considered in the discussion sections.
Referee cross-commenting
Reviewer 1:
Dear Reviewer #2, The idea of performing control experiments in the point-mutant Cx43 or KO/KI mouse makes sense. If you think this is essential, then please enter it into your overall comments. However, performing this experiment will not be easily done within the one month revision time frame you proposed. Cheers.
Reviewer 2:
I think the data of this manuscript is missing a control animal that would present all the compensation changes that occur during development that occur in mice carrying the mutated Cx43. Alternatively, a doable experiment would be the use of inducible KO/KI. When comparing susceptibility to any drug it is very important to count with the best control possible. Otherwise, the results cannot be interpreted as cause-effect response.
Reviewer 1:
I agree with reviewer #2 that adding those two references will improve the ms. For the second ref mentioned, the doi link did not work; does reviewer #2 mean this ref: https://doi.org/10.1073/pnas.0603154104? "Change in permeant size selectivity by phosphorylation of connexin 43 gap-junctional hemichannels by PKC
If completed and/or interpreted carefully it could be relevant to enrich our knowledge on the importance of glial Cx43 in epilepsy.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary
Muñoz-Ballester et al. investigated the effects of TBI on Cx43 expression and function following TBI. They have examined the potential role of Cx43-containing gap junctions (GJs) and/or hemichannels (HCs), in their phosphorylated and unphosphorylated forms, in the mouse cortex. The experiments and hypotheses are simple and direct, but the results are not strong and generally correlative.
Major comments
Minor comments
Referee cross-commenting
Reviewer 1:
Dear Reviewer #2, The idea of performing control experiments in the point-mutant Cx43 or KO/KI mouse makes sense. If you think this is essential, then please enter it into your overall comments. However, performing this experiment will not be easily done within the one month revision time frame you proposed. Cheers.
Reviewer 2:
I think the data of this manuscript is missing a control animal that would present all the compensation changes that occur during development that occur in mice carrying the mutated Cx43. Alternatively, a doable experiment would be the use of inducible KO/KI. When comparing susceptibility to any drug it is very important to count with the best control possible. Otherwise, the results cannot be interpreted as cause-effect response.
Reviewer 1:
I agree with reviewer #2 that adding those two references will improve the ms. For the second ref mentioned, the doi link did not work; does reviewer #2 mean this ref: https://doi.org/10.1073/pnas.0603154104? "Change in permeant size selectivity by phosphorylation of connexin 43 gap-junctional hemichannels by PKC
The strength of this study is using a single-point mutant mouse of the Cx43 to assess the role of Cx43 phosphorylation in TBI-induced seizure susceptibility, pinpointing the molecular target. One limitation is that, while the S368A mutant directly addresses the seizure susceptibility issue, pharmacological treatments like CBX and Tat-Gap19 do not test the effects of phosphorylation. Another weakness is that the key mechanism underlying the effects of TBI on Cx43 is still unclear. This is because TBI does not change Cx43 plaque size (Fig. 5), it alters EtBr dye uptake in cells that may or may not be astrocytes (Fig. 3), and it changes Cx43 solubility, but this is correlative for GJs vs HCs. The overall idea of Cx43 contributing to seizures and TBI is interesting for the general neuroscience community. However, this study can use more direct experimentation to support its hypothesis.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
Summary The authors focused on medaka retinal organoids to investigate the mechanism underlying the eye cup morphogenesis. The authors succeeded to induce lens formation in fish retinal organoids using 3D suspension culture with minimal growth factor-containing media containing the Hepes. At day 1, Rx3:H2B-GFP+ cells appear in the surface region of organoids. At day 1.5, Prox1+cells appear in the interface area between the organoid surface and the core of central cell mass, which develops a spherical-shaped lens later. So, Prox1+ cells covers the surface of the internal lens cell core. At day 2, foxe3:GFP+ cells appear in the Prox1+ area, where early lens fiber marker, LFC, starts to be expressed. In addition, foxe3:GFP+ cells show EdU+ incorporation, indicating that foxe3:GFP+ cells have lens epithelial cell-characters. At day 4, cry:EGFP+ cells differentiate inside the spherical lens core, whose the surface area consists of LFC+ and Prox1+ cells. Furthermore, at day 4, the lens core moves towards the surface of retinal organoids to form an eye-cup like structure, although this morphogenesis "inside out" mechanism is different from in vivo cellular "outside -in" mechanism of eye cup formation. From these data, the authors conclude that optic cup formation, especially the positioning of the lens, is established in retinal organoids though the different mechanism of in vivo morphogenesis.
Overall, manuscript presentation is nice. However, there are still obscure points to understand background mechanism. My comments are shown below.
Major comments 1) At the initial stage of retinal organoid morphogenesis, a spherical lens is centrally positioned inside the retinal organoids, by covering a central lens core by the outer cell sheet of retinal precursor cells. I wonder if the formation of this structure may be understood by differential cell adhesive activity or mechanical tension between lens core cells and retinal cell sheet, just like the previous study done by Heisenberg lab on the spatial patterning of endoderm, mesoderm and ectoderm (Nat. Cell Biol. 10, 429 - 436 (2008)). Lens core cells may be integrated inside retinal cell mass by cell sorting through the direct interaction between retinal cells and lens cells, or between lens cells and the culture media. After day 1, it is also possible to understand that lens core moves towards the surface of retinal organoids, if adhesive/tensile force states of lens core cells may be change by secretion of extracellular matrix. I wonder if the authors measure physical property, adhesive activity and solidness, of retinal precursor cells and lens core cells. If retinal organoids at day 1 are dissociated and cultured again, do they show the same patterning of internal lens core covering by the outer retinal cell sheet? *Response: The question, whether different adhesive activity is involved in cell sorting and lens formation is indeed very intriguing. To address this point, we will include additional experiment (see Revision Plan, experiment 1). This experiment will be based on the dissociation and re-aggregation of lens-forming organoids as suggested by the reviewer. To monitor cell type specific sorting, we will employ a lens progenitor reporter line Foxe3::GFP and the retina-specific Rx2::H2B-RFP. If different adhesive activities of lens and retinal progenitor cells are involved and drive the process of cell sorting, dissociation and re-aggregation will result in cell sorting based on their identity. *
2) Optic cup is evaginated from the lateral wall of neuroepithelium of the diencephalon. In zebrafish, cell movement occurs from the pigment epithelium to the neural retina during eye morphogenesis in an FGF-dependent manner. How the medaka optic cup morphogenesis is coordinated? I also wonder if the authors conduct the tracking of cell migration during optic cup morphogenesis to reveal how cell migration and cell division are regulated in lens of the Medaka retinal organoids. It is also interesting to examine how retinal cell movement is coordinated during Medaka retinal organoids. Response: Looking into the detail of how optic cup-looking tissue arrangement of ocular organoids is achieved on cellular level is of course interesting. Our previous study showed that optic vesicles of medaka retinal organoids do not form optic cups (for details please see Zilova et al., 2021, eLIFE). We assume that the formation of cup-looking structure of the ocular organoids is mediated by the following processes: establishment of retina and lens domains at the specific region of the organoid – retina on the surface and lens in the center (see Figure S2 d and Figure 3e, and Figure 4). Further dislocation of the centrally formed lens towards the organoid periphery through the retina layer, places the lens to the periphery while retinal cells stay static. We assume that the “cup-like” shape is acquired by extrusion of the lens from the center of the organoid. To clarify this process with respect to tissue rearrangements and cell movements, we will include additional experiments (see Revision Plan, experiment 2) and follow lens- and retina-fated cells (by employing lens-specific Foxe3::GFP and retina-specific Rx2::H2B-RFP reporter lines) through the process of lens extrusion to dissect individual contribution of retinal/lens cells to this process (cross-reference with Reviewer #2).
3) The authors showed that blockade of FGF signaling affects lens fiber differentiation in day 1-2, whereas lens formation seems to be intact in the presence of FGF receptor inhibitor in day 0-1. I suggest the authors to examine which tissue is a target of FGF signaling in retinal organoids, using markers such as pea3, which is a downstream target of ERK branch of FGF signaling. Since FGF signaling promotes cell proliferation, is the lens core size normal in SU5402-treated organoids from day 0 to day 1?
Response: Assessing the activity of FGF signaling (cross-reference to Reviewer #3) in the organoids is indeed an important point. To address which tissue is the target of FGF signaling we will include additional experiments and assess the phosphorylation status of ERK (pERK) and expression of the ERK downstream target pea3, as suggested by the reviewer (see Revision Plan, experiment 3). That will allow to identify the tissue within the organoid responding to the Fgf signaling.
Lens core size of organoids treated with SU5402 from day 0 to day 1 is fully comparable to the control (please see Figure 6b).
4) Fig. 3f and 3g indicate that there is some cell population located between foxe3:GFP+ cells and rx2:H2B-RFP+ cells. What kind of cell-type is occupied in the interface area between foxe3:GFP+ cells and rx2:H2B-RFP+ cells?
Response: That is for sure an interesting question. We are aware of this population of cells. We currently do not have data that would with certainty clarify the fate of those cells. We are currently following up on that question with the use of scRNA sequencing, however we will not be able to address this question in the current manuscript.* * 5) Fig. 5e indicates the depth of Rx3 expression at day 1. Is the depth the thickness of Rx3 expressing cell sheet, which covers the central lens core in the organoids? If so, I wonder if total cell number of Rx3 expressing cell sheet may be different in each seeded-cell number, because thickness is the same across each seeded-cell number, but the surface area size may be different depending on underneath the lens core size. Please clarify this point.
*Response: Yes. Figure 5e indicates the thickness of the cell sheet expressing Rx3 that lies on the surface of the organoid. Indeed, the number of Rx3-expressing cells (and lens cells) scales with the size of the organoid as stated in the submitted manuscript. *
6) Noggin application inhibits lens formation at day 0-1. BMP signaling regulates formation of lens placode and olfactory placode at the early stage of development. It is interesting to examine whether Noggin-treated organoid expands olfactory placode area. Please check forebrain territory markers.
Response: What tissue differentiates at the expense of the lens in BMP inhibitor-treated organoids is of course an intriguing question. To address the identity of cells differentiated under this condition we will include an additional experiment (see Revision Plan, experiment 4 as suggested by the reviewer). We will check for the expression of Lhx2, Otx2 and Huc/D to address this point.
I have no minor comments
**Referees cross-commenting**
I agree that all reviewers have similar suggestions, which are reasonable and provided the same estimated time for revision.
Reviewer #1 (Significance (Required)):
Strength: This study is unique. The authors examined eye cup morphogenesis using fish retinal organoids. Eye cup normally consists of the lens, the neural retina, pigment epithelium and optic stalk. However, retinal organoids seem to be simple and consists of two cell types, lens and retina. Interestingly, a similar optic cup-like structure is achieved in both cases; however, underlying mechanism is different. It is interesting to investigate how eye morphogenesis is regulated in retinal organoids,under the unconstrained embryo-free environment.
Limitation: Description is OK, but analysis is not much profound. It is necessary to apply a bit more molecular and cellular level analysis, such as tracking of cell movement and visualization of FGF signnaling in organoid tissues.
Advancement: The current study is descriptive. Need some conceptual advance, which impact cell biology field or medical science.
Audience: The target audience of current study are still within ophthalmology and neuroscience community people, maybe translational/clinical rather than basic biology. To beyond specific fields, need to formulate a general principle for cell and developmental biology.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
In this study from Stahl et al., the authors demonstrate that medaka pluripotent embryonic cells can self-organise into eye organoids containing both retina and lens tissues. While these organoids can self-organize into an eye structure that resembles the vertebrate eye, they are built from a fundamentally different morphogenetic process - an "inside-out" mechanism where the lens forms centrally and moves outward, rather than the normal "outside-in" embryonic process. This is a very interesting discovery, both for our understanding of developmental biology and the potential for tissue engineering applications. The study would benefit from some additional experiments and a few clarifications.
The authors suggest that the lens cells are the ones that move from the central to a more superficial position. Is this an active movement of lens cells or just the passive consequence of the retina cells acquiring a cup shape? Are the retina cells migrating behind the lens or the lens cells pushing outwards? High-resolution imaging of organoid cup formation, tracking retina cells in combination with membrane labeling of all cells would help elucidate the morphogenetic processes occurring in the organoids. Membrane labeling would also be useful as Prox1 positive lens cells appear elongated in embryos while in the organoids, cell shapes seem less organised, less compact and not elongated (for example as shown in Fig 3f,g).
Response: Looking into the detail of how optic cup-looking tissue arrangement of ocular organoids is achieved on cellular level is of course interesting. We assume that the formation of cup-looking structures of the ocular organoids is mediated by following processes: establishment of retina and lens domains at a specific region of the organoid – retina on the surface and lens in the center (see Figure S2 d and Figure 3e, and Figure 4). Further dislocation of centrally formed lenses towards the organoid periphery through the retina layer, place the lens to the periphery while retinal cells stay static. We assume that the “cup-like” shape is acquired by extrusion of the lens. To clarify this process with respect to tissue rearrangements and cell movements, we will include additional experiments (see Revision Plan, experiment 2). We will follow lens- and retina-fated cells (by employing lens-specific Foxe3::GFP and retina-specific Rx2::H2B-RFP reporter lines) through the process of lens extrusion to dissect the individual contribution of retinal/lens cells to this process (cross-reference with Reviewer #1).
The organoids could be a useful tool to address how cell fate is linked to cell shape acquisition. In the forming organoids, retinal tissue initially forms on the outside, while non-retinal tissue is located in the centre; this central tissue later expresses lens markers. Do the authors have any insights into why fate acquisition occurs in this pattern? Is there a difference in proliferation rates between the centrally located cells and the external ones? Could it be that highly proliferative cells give rise to neural retina (NR), while lower proliferating cells become lens? *Response: The question how is the retinal and lens domain established in this specific manner is indeed intriguing and very interesting. We dedicated a part of the discussion to this topic. We discuss the role of the diffusion limit and the potential contribution of BMB and FGF signaling to this arrangement. Additional experiments (see Revision Plan, experiment 3) addressing the source and target tissues of FGF and BMP signaling in the organoid will ultimately bring more clarity to our understanding of the tissue arrangements in the organoid. *
*Although analysis of the proliferation rate of the cells at the surface and in the central region of the organoid might possibly show some differences in the proliferation rates between lens and retinal cells, we do not have any indications, that the proliferation rate itself would be instructive or superior to the cell fate decisions. *
What happens in organoids that do not form lenses? Do these organoids still generate foxe3 positive cells that fail to develop into a proper lens structure? And in the absence of lens formation, does the retina still acquire a cup shape?
*Response: Lens formation is primarily dependent on acquisition/specification of Foxe3-expressing lens placode progenitors. If those are not present, a lens does not develop. Once Foxe3-expressing progenitors are established, a lens is formed in unperturbed conditions (measured by the presence of expression of crystallin proteins). In such conditions, organoids that do not have a lens, do not carry Foxe3-expressing cells. *
*In the absence of the lens, the organoid is composed of retinal neuroepithelium, that does not form an optic cup (for details of such phenotypes please see Zilova et al., 2021, eLIFE). *
The author suggest that lens formation occurs even in the absence of Matrigel. Is the process slower in these conditions? Are the resulting organoids smaller? While there are indeed some LFC expressing cells by day2, these cells are not very well organised and the pattern of expression seems dotty. Moreover, LFC staining seems to localise posterior to the LFC negative, lens-like structure (e.g. Fig.S1 3o'clock). How do these organoids develop beyond day 4? Do they maintain their structural integrity at later stages? The role of HEPES in promoting organoid formation is intriguing. Do the authors have any insights into why it is important in this context? Have the authors tried other culture conditions and does culture condition influence the morphogenetic pathways occurring within the organoids? *Response: We thank the reviewer for pointing this out. We were not clear in the wording and describing of our observation. Indeed, Matrigel is not required for acquisition of lens fate, which can be demonstrated with the expression of lens-specific markers. However, the presence of Matrigel has a profound impact on the structural aspects of organoid formation. Matrigel is essential for organization of retinal-committed cells into the retinal epithelium (Zilova et al., 2021, eLIFE). The absence of the structure of the retinal epithelium can indeed negatively impact on the cellular organization and the overall lens structure. To clarify the contribution of the Matrigel to the speed of organoid lens development and to the overall structure of the organoid lens we will perform additional experiments (see Revision Plan, experiment 5). With the use of Foxe3::GFP reporter line we will measure the onset of the lens-specific gene expression. In addition, we will use the immunohistochemistry to assess the gross morphology and size of the organoids grown without the Matrigel (cross-reference with Reviewer #3). *
*The role of the HEPES in lens formation is indeed very intriguing and currently under investigation. As HEPES is mainly used to regulate pH of the culture media and pH might have an impact on multiple cellular processes, it will require significant time investment to dissect molecular mechanism underlying the effect of HEPES on the process of lens formation (cross reference with Reviewer #3) and therefore cannot be addressed in the current manuscript. *
**Referees cross-commenting** Pleased to see that all the other reviewers are positive about the study and raise similar concerns and comments
Reviewer #2 (Significance (Required)):
This is a very interesting paper, and it will be important to determine whether this alternative morphogenetic process is specific to medaka or if similar developmental routes can be recapitulated in organoid cultures from other vertebrate species.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
Summary: The manuscript by Stahl and colleagues reports an approach to generate ocular organoids composed of retinal and lens structures, derived from Medaka blastula cells. The authors present a comprehensive characterisation of the timeline followed by lens and retinal progenitors, showing these have distinct origins, and that they recapitulate the expression of differentiation markers found in vivo. Despite this molecular recapitulation, morphogenesis is strikingly different, with lens progenitors arising at the centre of the organoid, and subsequently translocating to the outside.
Comments:
-The manuscript presents a beautiful set of high quality images showing expression of lens differentiation markers over time in the organoids. The set of experiments is very robust, with high numbers of organoids analysed and reproducible data. The mechanism by which lens specification is promoted in these organoids is, however, poorly analysed, and the reader does not get a clear understanding of what is different in these experiments, as compared to previous attempts, to support lens differentiation. There is a mention to HEPES supplementation, but no further analysis is provided, and the fact that the process is independent of ECM contradicts, as the authors point out, previous reports. The manuscript would benefit from a more detailed analysis of the mechanisms that lead to lens differentiation in this setting.
*Response: The role of the HEPES in lens formation is indeed very intriguing and under current investigation. As HEPES is mainly used to regulate pH of the culture media and pH might have an impact on multiple cellular processes it will require a significant time investment to dissect molecular mechanism underlying the effect of HEPES on the process of lens formation (cross reference with Reviewer #2) and therefore unfortunately cannot be addressed in the current manuscript. *
*To clarify the contribution of the Matrigel to the organoid lens development we will perform additional experiments (see Revision Plan, experiment 5). With the use of Foxe3::GFP reporter line we will measure the onset of the lens-specific gene expression. In addition, we will use the immunohistochemistry to assess the gross morphology and size of the organoids grown without the Matrigel (cross-reference with Reviewer #2). * -The markers analysed to show onset of lens differentiation in the organoids seem to start being expressed, in vivo, when the lens placode starts invaginating. An analysis of earlier stages is not presented. This would be very informative, allowing to determine whether progenitors differentiate as placode and neuroepithelium first, to subsequently continue differentiating into lens and retina, respectively. Could early placodal and anterior neural plate markers be analysed in the organoids? This would provide a more complete sequence of lens vs retina differentiation in this model.
Response: Yes. The figures show the expression of lens and retinal markers in the embryo in later developmental stages and the timing of their expression can be documented with higher temporal resolution. In the revised version of the manuscript, we will provide the information about the onset of expression of Rx3::H2B-GFP (retina) and Foxe3::GFP (lens) (see attached figure). Rx3 represents one of the earlies markers labeling the presumptive eye field within the region of the anterior neural plate (S16, late gastrula). FoxE3::GFP expression can be detected within the head surface ectoderm before the lens placode is formed showing that Foxe3 is a suitable marker of placodal progenitors in medaka.
*We are convinced that the onset of Rx3 and Foxe3-driven reporters is early enough to make the claim about the separate origin of the lens (placodal) and retinal (anterior neuroectoderm) tissues within the ocular organoids. *
-The analysis of BMP and Fgf requirement for lens formation and differentiation is suggestive, but the source of these signals is not resolved or mentioned in the manuscript. Are BMP4 and Fgf8 expressed by the organoids? Where are they coming from?
Response: Indeed, addressing the source of BMP and FGF activation would bring more clarity in understanding the mechanism of retina/lens specification within the ocular organoids (cross reference with Reviewer #1). To address this point, we will include additional experiments (see Revision Plan, experiment 3). We will analyze the expression of respective ligands (Bmp4 and Fgf8) and activation of downstream effectors of BMP and FGF signaling pathways within the ocular organoids as suggested by Reviewer #1 and Reviewer #3.
-The fact that the lens becomes specified in the centre of the organoid is striking, but it is for me difficult to visualise how it ends up being extruded from the organoid. Did the authors try to follow this process in movies? I understand that this may be technically challenging, but it would certainly help to understand the process that leads to the final organisation of retinal and lens tissues in the organoid. There is no discussion of why the morphogenetic mechanism is so different from the in vivo situation. The manuscript would benefit from explicitly discussing this. Response: Following the extruding lens in vivo is indeed very relevant suggestion. To clarify the process of ocular organoid formation in the respect of tissue rearrangements and cell movements, we will include additional experiment (see Revision Plan, experiment 2). We will follow lens- and retina-fated cells (by employing lens-specific Foxe3::GFP and retina-specific Rx2::H2B-RFP reporter lines) through the process of lens extrusion (cross-reference with Reviewer #1 and Reviewer #2).
**Referees cross-commenting**
We all seem to have similar comments and concerns. I think overall the suggestions are feasible and realistic for the timeframe provided.
Reviewer #3 (Significance (Required)):
This study describes a reproducible approach to differentiate ocular organoids composed of lens and retinal tissues. The characterisation of lens differentiation in this model is very detailed, and despite the morphogenetic differences, the molecular mechanisms show many similarities to the in vivo situation. The manuscript however does not highlight, in my opinion, why this model may be relevant. Clearly articulating this relevance, particularly in the discussion, will enhance the study and provide more clarity to the readers regarding the significance of the study for the field of organoid research, ocular research and regenerative studies.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary: The manuscript by Stahl and colleagues reports an approach to generate ocular organoids composed of retinal and lens structures, derived from Medaka blastula cells. The authors present a comprehensive characterisation of the timeline followed by lens and retinal progenitors, showing these have distinct origins, and that they recapitulate the expression of differentiation markers found in vivo. Despite this molecular recapitulation, morphogenesis is strikingly different, with lens progenitors arising at the centre of the organoid, and subsequently translocating to the outside.
Comments:
Referees cross-commenting
We all seem to have similar comments and concerns. I think overall the suggestions are feasible and realistic for the timeframe provided.
This study describes a reproducible approach to differentiate ocular organoids composed of lens and retinal tissues. The characterisation of lens differentiation in this model is very detailed, and despite the morphogenetic differences, the molecular mechanisms show many similarities to the in vivo situation. The manuscript however does not highlight, in my opinion, why this model may be relevant. Clearly articulating this relevance, particularly in the discussion, will enhance the study and provide more clarity to the readers regarding the significance of the study for the field of organoid research, ocular research and regenerative studies.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this study from Stahl et al., the authors demonstrate that medaka pluripotent embryonic cells can self-organise into eye organoids containing both retina and lens tissues. While these organoids can self-organize into an eye structure that resembles the vertebrate eye, they are built from a fundamentally different morphogenetic process - an "inside-out" mechanism where the lens forms centrally and moves outward, rather than the normal "outside-in" embryonic process. This is a very interesting discovery, both for our understanding of developmental biology and the potential for tissue engineering applications. The study would benefit from some additional experiments and a few clarifications. The authors suggest that the lens cells are the ones that move from the central to a more superficial position. Is this an active movement of lens cells or just the passive consequence of the retina cells acquiring a cup shape? Are the retina cells migrating behind the lens or the lens cells pushing outwards? High-resolution imaging of organoid cup formation, tracking retina cells in combination with membrane labeling of all cells would help elucidate the morphogenetic processes occurring in the organoids. Membrane labeling would also be useful as Prox1 positive lens cells appear elongated in embryos while in the organoids, cell shapes seem less organised, less compact and not elongated (for example as shown in Fig 3f,g). The organoids could be a useful tool to address how cell fate is linked to cell shape acquisition. In the forming organoids, retinal tissue initially forms on the outside, while non-retinal tissue is located in the centre; this central tissue later expresses lens markers. Do the authors have any insights into why fate acquisition occurs in this pattern? Is there a difference in proliferation rates between the centrally located cells and the external ones? Could it be that highly proliferative cells give rise to neural retina (NR), while lower proliferating cells become lens?
What happens in organoids that do not form lenses? Do these organoids still generate foxe3 positive cells that fail to develop into a proper lens structure? And in the absence of lens formation, does the retina still acquire a cup shape?
The author suggest that lens formation occurs even in the absence of Matrigel. Is the process slower in these conditions? Are the resulting organoids smaller? While there are indeed some LFC expressing cells by day2, these cells are not very well organised and the pattern of expression seems dotty. Moreover, LFC staining seems to localise posterior to the LFC negative, lens-like structure (e.g. Fig.S1 3o'clock). How do these organoids develop beyond day 4? Do they maintain their structural integrity at later stages? The role of HEPES in promoting organoid formation is intriguing. Do the authors have any insights into why it is important in this context? Have the authors tried other culture conditions and does culture condition influence the morphogenetic pathways occurring within the organoids?
Referees cross-commenting
Pleased to see that all the other reviewers are positive about the study and raise similar concerns and comments
This is a very interesting paper, and it will be important to determine whether this alternative morphogenetic process is specific to medaka or if similar developmental routes can be recapitulated in organoid cultures from other vertebrate species.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary
The authors focused on medaka retinal organoids to investigate the mechanism underlying the eye cup morphogenesis. The authors succeeded to induce lens formation in fish retinal organoids using 3D suspension culture with minimal growth factor-containing media containing the Hepes. At day 1, Rx3:H2B-GFP+ cells appear in the surface region of organoids. At day 1.5, Prox1+cells appear in the interface area between the organoid surface and the core of central cell mass, which develops a spherical-shaped lens later. So, Prox1+ cells covers the surface of the internal lens cell core. At day 2, foxe3:GFP+ cells appear in the Prox1+ area, where early lens fiber marker, LFC, starts to be expressed. In addition, foxe3:GFP+ cells show EdU+ incorporation, indicating that foxe3:GFP+ cells have lens epithelial cell-characters. At day 4, cry:EGFP+ cells differentiate inside the spherical lens core, whose the surface area consists of LFC+ and Prox1+ cells. Furthermore, at day 4, the lens core moves towards the surface of retinal organoids to form an eye-cup like structure, although this morphogenesis "inside out" mechanism is different from in vivo cellular "outside -in" mechanism of eye cup formation. From these data, the authors conclude that optic cup formation, especially the positioning of the lens, is established in retinal organoids though the different mechanism of in vivo morphogenesis.
Overall, manuscript presentation is nice. However, there are still obscure points to understand background mechanism. My comments are shown below.
Major comments
I have no minor comments
Referees cross-commenting
I agree that all reviewers have similar suggestions, which are reasonable and provided the same estimated time for revision.
Strength: This study is unique. The authors examined eye cup morphogenesis using fish retinal organoids. Eye cup normally consists of the lens, the neural retina, pigment epithelium and optic stalk. However, retinal organoids seem to be simple and consists of two cell types, lens and retina. Interestingly, a similar optic cup-like structure is achieved in both cases; however, underlying mechanism is different. It is interesting to investigate how eye morphogenesis is regulated in retinal organoids,under the unconstrained embryo-free environment.
Limitation: Description is OK, but analysis is not much profound. It is necessary to apply a bit more molecular and cellular level analysis, such as tracking of cell movement and visualization of FGF signnaling in organoid tissues.
Advancement: The current study is descriptive. Need some conceptual advance, which impact cell biology field or medical science.
Audience: The target audience of current study are still within ophthalmology and neuroscience community people, maybe translational/clinical rather than basic biology. To beyond specific fields, need to formulate a general principle for cell and developmental biology.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
__We thank the reviewers for the supportive suggestions and comments. We have addressed all comments underneath the original text in red. As suggested, we added to line numbers to the text and use these numbers to refer to the changes made. __
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
The manuscript is well written and presents solid data, most of which is statistically analyzed and sound. Given that the author's previous comprehensive publications on seipin organization and interactions, it might be beneficial (particularly in the title and abstract) to emphasize that this manuscript focuses on the metabolic regulation of lipid droplet assembly by Ldb16, to distinguish it from previous work. Perhaps one consideration, potentially interesting, involves changes in lipid droplet formation under the growth conditions used for galactose-mediated gene induction.
We thank the reviewer for the supportive comments and suggestions.
Comments: (1) Fig. 3 and 4. The galactose induction of lipid droplet biogenesis in are1∆/2∆ dga1∆ lro1∆ cells though activation of a GAL1 promoter fusion to DGA1 is a sound approach for regulating lipid droplet formation. Although unlikely, carbon sources can impact lipid droplet proliferation and (potentially interesting) metabolic changes under growth in non-fermentable carbon sources may impact lipid droplet biogenesis; in fact, oleate has significant effects (e.g. PMID: 21422231; PMID: 21820081). The GAL1 promoter is a very strong promoter and the overexpression of DGA1 via this heterologous promoter might itself cause unforeseen changes. Affirmation of the results using another induction system might be beneficial.
We thank the reviewer for these suggestions. In this study we focused on the organisation of the yeast seipin complex during the process of LD formation. We chose to use galactose-based induction of Dga1 because this is a well-established and widely used assay in the field, extensively characterized by many groups over the years. The tight control it provides, enabling synchronous and rapid LD induction, makes it the method of choice for many researchers. Importantly, the LDs formed using this assay are morphologically normal and involve the same components as LDs formed under other conditions.
Regarding the role of metabolism in LD formation, it is worth noting that galactose is metabolized by yeast primarily through fermentation, following its conversion to UDP-glucose. Therefore, its use does not involve drastic metabolic changes. The impact of metabolism in LD biogenesis is an interesting question but it falls beyond the scope of the current study.
(2) Fig. 3B. Although only representative images are shown, the panel convincingly shows that lipid droplets do form upon galactose induction of DGA1 in are1∆/2∆ dga1∆ lro1∆ cells. However, it does not show to what extent. Are lipid droplets synthesized at WT levels? How many cells were counted? How many lipid droplets per cell? Is there a statistical difference with respect to WT cells?
We did not assess these parameters in this study. The aim of the study was to assess the relations between components of the seipin complex with and without lipid droplets. For this purpose, inducing lipid droplet formation over a 4-hour period was sufficient to address that specific question. As mentioned above, LDs formed using this assay are morphologically normal and involve the same components as LDs formed under other conditions. This being said, it is known that prolonged overexpression of Dga1 (> 12hours) can lead to enlarged LDs.
(3) Fig. 2D. It is not clear how standard deviation can be meaningfully applied to two data points, let alone providing a p-value. For some of these experiments, triplicate trials might provide a more robust statistical sampling.
We thank the reviewer for this suggestion. We have added 2 more repeats to the Co-IP in figure 2.
Reviewer #1 (Significance (Required)):
Klug and Carvalho report on the lipid droplet architecture of the yeast seipin complex. Specifically, the mechanism of yeast seipin Sei1 binding to Ldo16 and the subsequent recruitment of Ldb45 is analyzed. These results follow from a recent publication (PMID: 34625558) from the same authors and aims to define a more precise role for the components of the seipin complex. Using photo-crosslinking, Ldo45 and Ldo16 interactions are analyzed in the context of lipid droplet assembly.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
Summary:
Klug and Carvalho apply a photo-crosslinking approach, which has been extensively used in the Carvalho group, to investigate the subunit interactions of the seipin complex in yeast. The authors apply this approach to further study possible changes within the seipin complex following induction of neutral lipid synthesis and lipid droplet (LD) formation. The authors propose that Ldo45 makes contact with Ldb16 and that the seipin complex subunits assemble even in the absence of LDs.
Major comments:
Overall, this is a focused and well-executed study on one of the fundamental structural components of LDs. The study addresses the subunit interactions of the seipin complex but does not look into their functional consequences, for example how the mutations on Ldb16 that affect its interaction with Ldo45, influence LD formation; similarly, the authors make the interesting observation that Ldo16 may be differentially affected by the lack of neutral lipids (Fig. 3A) but this observation is not explored.
We thank the reviewer for this comment. The Ldb16 mutations analyzed in this study have been previously characterized by us (see Klug et al., 2021 – Figure 3) and exhibit a mild defect in lipid droplet (LD) formation. This phenotype is unlikely to result from impaired Ldo16/45 recruitment, as deletion of Ldo proteins causes only a very mild effect on LD formation (as shown in Teixeira et al., 2018 and Eisenberg-Bord et al., 2018).
We agree that the differential effect on Ldo proteins by the absence of neutral lipids is particularly interesting. However, its exploration falls outside of the scope of the current study and should be thoroughly investigated in the future.
For the crosslinking pull-downs (Fig. 1), it seems that the authors significantly overexpress (ADH1 promoter) the Ldb16 subunit that carries the various photoreactive amino acid residues, while keeping the other (tagged) seipin complex members at endogenous levels. Would not this imbalance affect the assembly of the complex and therefore the association of the different subunits with each other?
We thank the reviewer for this comment. The in vivo site-specific crosslinking is highly sensitive methodology to detect protein-protein interactions in a position-dependent manner. However, one of the caveats of the approach is the low efficiency of amber stop codon suppression and BPA incorporation. To mitigate this limitation, we (and others) induce the expression of the amber-containing protein (in this case Ldb16) from a strong constitutive promoter such as ADH1. Therefore, despite using a strong promoter, the overall levels of LDB16 remain comparable to endogenous levels due to the inherently low efficiency of amber suppression. Moreover, it is known that when not bound to Sei1, Ldb16 is rapidly degraded in a proteasome dependent manner (Wang, C.W. 2014), further preventing its accumulation.
Although the authors do show delta4 cells with no LDs (Fig. 3B, 0h), galactose-inducible systems in yeast are known to be leaky. Given that the authors' conclusion that the complex is "pre-assembled" irrespective of the addition of galactose, I think it would be important to confirm biochemically that there is no neutral lipid at time point 0. Alternatively, it may be better to simply compare wt vs dga1 lro1 or are1are2 mutants - there is no need for GAL induction since the authors look at one time point only.
Among the various regulable promoters, GAL1 shows a superior level of control. For example, expression of essential genes from GAL1 promoter frequently leads to cell death in glucose containing media, a condition that represses GAL1 promoter. Having said this, we cannot exclude that minute amounts of DGA1 are expressed prior galactose induction. However, if this is the case, the resulting levels of TAG are insufficient to be detected by sensitive lipid dyes and to induce LDs, as noted by the reviewer. Therefore, we believe our conclusions remain valid. This is consistent that we use in the text, where we refer to LD formation rather than complete loss of neutral lipids. To make this absolutely clear we replaced the word “presence” to “abundance” in line 236.
Lastly, we do not agree with the reviewer that using double mutants (are1/2 or dga1/lro1 mutants) would be sufficient since these mutations are not sufficient to abolish LD formation – a key aspect of this study. The GAL1 system allows us to monitor 2 time points in the same cells –no LDs (time 0h) and with LDs (Time 4h). The system proposed by the reviewer would only allow a snap shot of steady state levels in different cells rather than within the same cell culture.
Some methodological issues could be better detailed. For example, which of the three delta4 strains was used to induce neutral lipid in Fig. 4B? How exactly were the quantifications in Fig. 4D performed (I assume they were done under non-saturating band intensity conditions, as for some residues it is difficult to conclude whether the blot aligns with the quantification results).
We thank the reviewer for these comments. We have clarified the strain number in the figure legend of figure 4B (strain yPC12630).
We have also added the following text in rows 437-441 in the methods section: “Reactive bands were detected by ECL (Western Lightning ECL Pro, Perkin Elmer #NEL121001EA), and visualized using an Amersham Imager 600 (GE Healthcare Life Sciences). Data quantification was performed using Image Studio software (Li-Cor) to measure line intensity under non saturating conditions.”
"our findings support the notion that Ldo45 is important for early steps of LD formation as previously proposed" I find this statement confusing given that the authors claim that Ldo45 is already bound to the complex before LD formation.
We thank the reviewer for raising this important point. We believe that our findings support previous hypotheses on the role of Ldo45. It has been suggested that Ldo45 is important for the early stages of lipid droplet (LD) formation (Teixeira et al., 2018; Eisenberg-Bord et al., 2018). As such, Ldo45 would need to be recruited to the seipin complex before or at the onset of LD formation. The observation that Ldo45 is present at the complex prior to LD formation provides strong support for its role in the initial steps of this process.
To clarify this idea in the manuscript, we have revised the sentence on line 310 as follows:
“Irrespective of the mechanism, our findings support the notion that Ldo45 plays a role in the early steps of LD formation, as previously proposed…”
The model in Fig. 5 is essentially the same as the one shown in Fig. 1G.
To aid the reader and avoid confusion, we intentionally used a similar color scheme throughout the manuscript. This may contribute to the perception that the figures are very similar. However, there are clear distinctions between them. In Figure 1G, we summarize our findings regarding the positioning of Ldo45 within the complex and note that we do not yet have data on Ldo16. Building upon these findings, in Figure 5 we speculate where Ldo16 might interact with Ldb16 and highlight that recruitment of both Ldo16 and Ldo45 increases with neutral lipid availability.
Therefore, we believe that both figures serve distinct and complementary purposes, and that each is useful for communicating our overall message.
Minor comments
In the pull-downs in Fig. 2C, it seems that full-length Ldb16 is not enriched after the FLAG IP. What is the reason of this?
We thank the reviewer for raising this interesting aspect. We do not know why this occurs, but it is clear that full length Ldb16 is not efficiently pulled down. We could speculate that this has to do with access to the FLAG moiety at the C terminus that may become inaccessible due to interactions or folding in the long unstructured C-terminus of Ldb16. This might explain why when we truncate the C terminus in the 1-133 mutant we achieve a more efficient IP.
At the blots at Fig. 2C and 3A, the anti-Dpm1 Ab seems to recognize in the IP fractions a band labelled as non-specific, however this band is absent from the input.
We thank the reviewer for raising this. This non-specific band is the light chain of the antibody used in the pull down that detaches from the matrix during elution – thus not found in the input. This is a common non-specific band that appears in Co-IP blots.
Reviewer #2 (Significance (Required)):
Regulation of seipin function is essential for proper LD biogenesis in eukaryotes, so this study addresses a fundamental question in the field. As stated above some functional analysis that goes beyond the biochemistry would be beneficial. There is some overlap with a recently published paper from the Wang group that also examines the assembly of seipin in yeast.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
The manuscript by Klug and Carvalho investigates the interaction of the yeast seipin complex (Sei1 and Ldb16) with Ldo45 and Ldo16. Using a site-specific photocrosslinking approach, the authors map some residues of the seipin complex in contact Ldo45, demonstrating that Ldo45 likely binds to Ldb16 in the center of the Sei1-Ldb16 complex. They find that both Ldo45 and Ldo16 copurify with Ldb16. Complex assembly is demonstrated to occur independently of the presence of neutral lipids. An Ldb16 mutant, harbouring the transmembrane domain (1-133) but lacking the cytosolic region (previously shown to allow normal LD formation and still bind to Sei1) showed photocrosslinks with Ldo45, but not Ldo16. No crosslinks between Sei1 and either Ldo45 or Ldo16 were detected.
Major: 1. Figure 2 shows CoIPs using different Ldb16 mutants/truncations to test for binding of Ldo45 and Ldo16. Both Ldo16 and Ldo45 copurify with full length Ldb16. Loss of the cytosolic part of Ldb16 strongly reduced binding of both Ldo45 and Ldo16, indicating that the TM-Helix-TM domain of Ldb16 (1-133) alone is not sufficient for proper binding of Ldo45 or Ldo16. The quantifications (2D and 2E) presented for this CoIP represent a n=2 with mean, standard deviation and statistics. To be a meaningful statistical analysis, the authors need to increase their n to at least n=3. In addition, they refer to the statistics they use here as "two-sided Fischer's T-test" in the respective Figure legend. To my knowledge, there is no such test, either it is Student's T-test or Fischer's exact test? Can the authors please clarify?
We thank the reviewer for this comment and suggestions. We have now included 2 additional repeats for this experiment and the results essentially support our conclusion.
The two-sided Fischer’s T-test is the name of the test in Graphpad- Prism. We wanted to acknowledge the test name so that the reader can trace the exact test we used in the program.
- Figure 2E shows the same data as 2D with different normalization to highlight the differences between binding to the domain 1-133 per se and binding to this domain when the linker helix is mutated. These mutations seem to cause a further decrease in binding of both Ldo45 and Ldo16. Still, effects are rather small, and the n=2 does not allow any meaningful statistical tests. To make this point, the authors should increase their sample number (at least n=3) to show that this difference is indeed meaningful and to allow statistical analysis.
We thank the reviewer for this comment and suggestions. We have now included 2 additional repeats for this experiment and the results essentially support our conclusion.
For Ldo16, no crosslinks were detected with Ldb16 TM-HelixTM domain (Figure 1). In line, CoIP demonstrated that the interaction between Ldo16 and Ldb16 was strongly reduced when the Ldb16 domain 1-133 was used for IP. Still, additional mutation of the linker helix in this 1-133 domain further reduced this interaction (to a similar extend as for Ldo45). Could the authors please clarify why the additional mutations in the linker helix region also decreased the binding of Ldo16, though the authors conclude from their crosslinking approach in Fig. 1 that Ldo16 does not interact with this region?
We thank the reviewer for raising this point. Our negative crosslinking results for Ldo16 do not exclude the possibility of binding to that region; rather, they indicate that we were unable to detect Ldo16 there. Additionally, mutations in the linker helix may influence how Ldb16 interacts with seipin, including its positioning within the seipin ring and the membrane bilayer. These structural changes could, in turn, affect Ldo16 recruitment in ways that we do not fully understand.
Similarly, also in 4D, a quantification with n=2 is presented, showing that some of the crosslinks are more prominently detectable when LD biogenesis is induced. The findings of this manuscript are completely based on results obtained with CoIP and photocrosslinking, and quantification of a sufficient n to allow statistical analysis will be essential.
While we agree that additional experiments are useful for the Co-IP because of variability between experiments, this is less of a concern for the photocrosslinking experiments. In the case of photocrosslinking, we typically see much less variability and normally, for a given position, the effects are much more “black and white”- either there is a crosslink or not.
Why is there nowhere a blot with crosslinked Ldb16 bands shown (but only non-crosslinked Ldb16, e.g. Fig. 1C)?
We thank the reviewer for this comment. In all cases the amount of crosslinked product is very minor. This is particularly obvious in the case of Ldb16, where the non-crosslinked species dominates in the blots (as can be observed in figure S1B).
Figure 3: The authors conclude that galactose-induced expression of either Dga1, Lro1 or Are1 in cells lacking all four enzymes for neutral lipid synthesis (quadruple deletion mutant) increases the levels of Ldb16. However, I do not see any difference on the FLAG-Ldb16 blot when comparing Ldb16 levels in the quadruple deletion mutant with or without Dga1, Lro1 or Are1, and no quantification is presented that might reveal very subtle differences not visible on the blot.
We agree with the reviewer and modified the text to more accurately describe our results.
OPTIONAL: Have the authors considered to assess which sites/domains of Ldo45 and Ldo16 are employed to bind to Ldb16?
This is a logical next step that will be undertaken in a future study.
Minor: 1. Page numbers would have been helpful to refer to specific text sections.
Page numbers have been added
- Figure 3C: Unclear to me why the authors label a part of their immunoblot where they detected HA with OSW5?
This was a mistake and has been corrected
- Figure 4D and corresponding figure legend could be improved in respect to labeling to clarify.
we have added an X axis label and made extra clarifications in the legend
- Please correct his sentence: "These variants we expressed in cells where the other subunits of the Sei1 complex were epitope tagged to facilitate detection and expressed their endogenous loci."
This sentence has been corrected
Reviewer #3 (Significance (Required)):
This is a short and interesting study completely based on UV-induced site-specific photocrosslinking and CoIPs that provides some new insights into the interaction surface between the Seipin complex and Ldo45 and the interaction between Ldo16 and Ldb16. Though in parts still premature, these findings will likely be of interest to the large community interested in lipid metabolism, expanding the role of Ldb16 from neutral lipid binding to regulator recruitment.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript by Klug and Carvalho investigates the interaction of the yeast seipin complex (Sei1 and Ldb16) with Ldo45 and Ldo16. Using a site-specific photocrosslinking approach, the authors map some residues of the seipin complex in contact Ldo45, demonstrating that Ldo45 likely binds to Ldb16 in the center of the Sei1-Ldb16 complex. They find that both Ldo45 and Ldo16 copurify with Ldb16. Complex assembly is demonstrated to occur independently of the presence of neutral lipids. An Ldb16 mutant, harbouring the transmembrane domain (1-133) but lacking the cytosolic region (previously shown to allow normal LD formation and still bind to Sei1) showed photocrosslinks with Ldo45, but not Ldo16. No crosslinks between Sei1 and either Ldo45 or Ldo16 were detected.
Major:
OPTIONAL: Have the authors considered to assess which sites/domains of Ldo45 and Ldo16 are employed to bind to Ldb16?
Minor:
This is a short and interesting study completely based on UV-induced site-specific photocrosslinking and CoIPs that provides some new insights into the interaction surface between the Seipin complex and Ldo45 and the interaction between Ldo16 and Ldb16. Though in parts still premature, these findings will likely be of interest to the large community interested in lipid metabolism, expanding the role of Ldb16 from neutral lipid binding to regulator recruitment.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
Klug and Carvalho apply a photo-crosslinking approach, which has been extensively used in the Carvalho group, to investigate the subunit interactions of the seipin complex in yeast. The authors apply this approach to further study possible changes within the seipin complex following induction of neutral lipid synthesis and lipid droplet (LD) formation. The authors propose that Ldo45 makes contact with Ldb16 and that the seipin complex subunits assemble even in the absence of LDs.
Major comments:
Overall, this is a focused and well-executed study on one of the fundamental structural components of LDs. The study addresses the subunit interactions of the seipin complex but does not look into their functional consequences, for example how the mutations on Ldb16 that affect its interaction with Ldo45, influence LD formation; similarly, the authors make the interesting observation that Ldo16 may be differentially affected by the lack of neutral lipids (Fig. 3A) but this observation is not explored.
Minor comments
In the pull-downs in Fig. 2C, it seems that full-length Ldb16 is not enriched after the FLAG IP. What is the reason of this?
At the blots at Fig. 2C and 3A, the anti-Dpm1 Ab seems to recognize in the IP fractions a band labelled as non-specific, however this band is absent from the input.
Regulation of seipin function is essential for proper LD biogenesis in eukaryotes, so this study addresses a fundamental question in the field. As stated above some functional analysis that goes beyond the biochemistry would be beneficial. There is some overlap with a recently published paper from the Wang group that also examines the assembly of seipin in yeast.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript is well written and presents solid data, most of which is statistically analyzed and sound. Given that the author's previous comprehensive publications on seipin organization and interactions, it might be beneficial (particularly in the title and abstract) to emphasize that this manuscript focuses on the metabolic regulation of lipid droplet assembly by Ldb16, to distinguish it from previous work. Perhaps one consideration, potentially interesting, involves changes in lipid droplet formation under the growth conditions used for galactose-mediated gene induction.
Comments:
Klug and Carvalho report on the lipid droplet architecture of the yeast seipin complex. Specifically, the mechanism of yeast seipin Sei1 binding to Ldo16 and the subsequent recruitment of Ldb45 is analyzed. These results follow from a recent publication (PMID: 34625558) from the same authors and aims to define a more precise role for the components of the seipin complex. Using photo-crosslinking, Ldo45 and Ldo16 interactions are analyzed in the context of lipid droplet assembly.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
1. Response to reviewers
We would like to thank the reviewers for carefully reading our manuscript and for their valuable comments in support for the publication of our investigation of rapid promoter evolution of accessory gland genes between Drosophila species and hybrids. We are glad to read that the reviewers find our work interesting and that it provides valuable insights into the regulation and divergence of genes through their promoters. We are encouraged by their acknowledgement of the overall quality of the work and the importance of our analyses in advancing the understanding of cis-regulatory changes in species divergence.
2. Point-by-point description of the revisions
Reviewer #
Reviewer Comment
Author Response/Revision
Reviewer 1
The authors test the hypothesis that promoters of genes involved in insect accessory glands evolved more rapidly than other genes in the genome. They test this using a number of computational and experimental approaches, looking at different species within the Drosophila melanogaster complex. The authors find an increased amount of sequence divergence in promoters of accessory gland proteins. They show that the expression levels of these proteins are more variable among species than randomly selected proteins. Finally, they show that within interspecific hybrids, each copy of the gene maintains its species-specific expression level.
We thank Reviewer 1 for their detailed review and positive feedback on our manuscript, and for their helpful suggestions. We have now fully addressed the points raised by Reviewer 1 and have provided the suggested clarifications and revisions to improve the flow, readability, and presentation of the data, which we believe have improved the manuscript significantly.
The work is done with expected standards of controls and analyses. The claims are supported by the analysis. My main criticism of the manuscript has to do not with the experiments or conclusion themselves but with the presentation. The manuscript is just not very well written, and following the logic of the arguments and results is challenging.
The problem begins with the Abstract, which is representative of the general problems with the manuscript. The Abstract begins with general statements about the evolution of seminal fluid proteins, but then jumps to accessory glands and hybrids, without clarifying what taxon is being studied, and what hybrids they are talking about. Then, the acronym Acp is introduced without explanation. The last two sentences of the Abstract are very cumbersome and one has to reread them to understand how they link to the beginning of the Abstract.
More generally, if this reviewer is to be seen as an "average reader" of the paper, I really struggled through reading it, and did not understand many of the arguments or rationale until the second read-through, after I had already read the bottom line. The paragraph spanning lines 71-83 is another case in point. It is composed of a series of very strongly worded sentences, almost all starting with a modifier (unexpectedly, interestingly, moreover), and supported by citations, but the logical flow doesn't work. Again, reading the paragraph after I knew where the paper was going was clearer, but on a first read, it was just a list of disjointed statements.
Since most of the citations are from the authors' own work, I suspect they are assuming too much prior understanding on the part of the reader. I am sure that if the authors read through the manuscript again, trying to look through the eyes of an external reader, they will easily be able to improve the flow and readability of the text.
We thank the reviewer for their detailed feedback and are glad that they acknowledge our work fully supports the claims of our manuscript. We also appreciate their helpful suggestions for improving the readability of the manuscript and have done our best to re-write the abstract and main text where indicated. In particular, the paragraph between lines 71-83 have been rewritten and we have taken care to write to non-expert readers.
1) In the analysis of expression level differences, it is not clear what specific stage / tissue the levels taken from the literature refer to. Could it be that the source of the data is from a stage or tissue where seminar fluid proteins will be expressed with higher variability in general (not just inter-specifically) and this could be skewing the results? Please add more information on the original source of the data and provide support for their validity for this type of comparison.
These were taken from publicly available adult male Drosophila datasets, listed in the data availability statement and throughout the manuscript. We have provided more detail on the tissue used for analysis of Acp gene expression levels.
2) The sentence spanning lines 155-157 needs more context.
We have added more context to lines 155-157.
3) Line 203-204: What are multi-choice enhancers?
We replaced the sentence with "... such as rapidly evolving enhancers or nested epistasis enhancer networks"
4) Figure 1: The terminology the authors use, comparing the gene of interest to "Genome" is very confusing. They are not comparing to the entire genome but to all genes in the genome, which is not the same.
We have changed the word "genome" to "all genes in the genome" on the reviewer's suggestion.
5) Figure 2: Changes between X vs. Y is redundant (either changes between X and Y or changes in X vs. Y).
We assume that the reviewer is referring to Fig. 2B, which does not measure changes between X and Y, but changes in distribution between Acps and the control group. We have explained this in the figure legend.
The manuscript addresses a general question in evolutionary biology - do control regions diverge more quickly protein coding regions. The answer is that yes, they do, but this is actually not very surprising. The work is probably thus of more interest to people interested in the copulatory proteins or in the evolution of mating systems, than to people interested in broader evolutionary questions.
We appreciate this reviewer's recognition of the significance of our work and would like to point out that there are very few studies looking at promoter evolution as detailed in the introduction. Of particular relevance, our study using Acp genes allows us to directly test the impact of promoter mutations on the expression by comparing two alleles in male accessory glands of Drosophila hybrids. Male accessory glands consist of only two secretory cell types allowing us to study evolution of gene expression in a single cell type (Acps are either expressed in main cells or secondary cells). Amid this unique experimental set up we can conclude that promoter mutations can act dominant, in contrast to mutations in protein coding regions, which are generally recessive. Thus, our study is unique in pointing out a largely overseen aspect of gene evolution.
Reviewer 2
This manuscript explores promoter evolution of genes encoding seminal fluid proteins expressed in the male accessory gland of Drosophila and finds cis-regulatory changes underlie expression differences between species. Although these genes evolve rapidly it appears that the coding regions rarely show signs of positive selection inferring that changes in their expression and hence promoter sequences can underlie the evolution of their roles within and among species.
We thank Reviewer 2 for their thorough review, positive feedback on the importance of our work, and suggestions for improving the manuscript. We have addressed all points raised by the reviewer, including analysis of Acp coding region evolution, additional analyses of hybrid expression data, and improved the clarity of the text.
Figure 1 illustrates evidence that the promoter regions of these gene have accumulated more changes than other sampled genes from the Drosophila genome. While this convinces that the region upstream of the transcription start site has diverged considerably in sequence (grey line compared to black line), Figure 1A also suggests the "Genespan" region which includes the 5'UTR but presumably also part of the coding region is also highly diverged. It would be useful to see how the pattern extends into the coding region further to compare further to the promoter region (although Fig 1H does illustrate this more convincingly).
The reviewer raises an interesting point, and certainly all parts of genes evolve. Fig. 1A shows the evolutionary rates of Acps compared to the genome average from phyloP27way scores calculated from 27 insect species. Since these species are quite distant it is unsurprising that they show divergence in coding regions as well as promoter regions. In fact, we addressed whether promoter regions evolve fast in closely related Drosophila species in Fig. 1H compared to coding regions. We have included an additional analysis of coding region evolution in Figure 1B.
Figure 2 presents evidence for significant changes in (presumably levels of) expression of male accessory gland protein (AcP) genes and ribosomal proteins genes between pairs of species, which is reflected in the skew of expression compared to randomly selected genes.
Correct, we have rephrased the statement for clarity.
Figure 3 shows detailed analysis for 3 selected AcP genes with significantly diverged expression. The authors claim this shows 'substitution' hotspots in the promoter regions of all 3 genes but this could be better illustrated by extending the plots in B-D further upstream and downstream to compare to these regions.
We picked the 300-nucleotide promoter region for this analysis as it accumulated significant changes as shown in Fig. 1E-H, and extending the G plots (Fig. 3B-D) to regions with lower numbers of sequence changes would not substantially change the conclusion. Specifically, this analysis identifies sequence change hotspots within fast-evolving promoter regions, rather than comparing promoter regions to other genomic regions, as we previously addressed. The plot is based on a cumulative distribution function and the significant positive slope in the upstream region where promoters are located identifies a hotspot for accumulation of substitutions. There could be other hotspots, but the point being made is that significant hotspots consistently appear in the promoter region of these three genes.
Figure 4 shows the results of expression analysis in parental lines of each pair of species and F1 hybrids. However the results are very difficult to follow in the figure and in the relevant text. While the schemes in A, C. E and G are helpful, the gel images are not the best quality and interpretations confusing. An additional scheme is needed to illustrate hypothetical outcomes of trans change, cis change and transvection to help interpret the gels. On line 169 (presumably referring to panels D and F although C and D are cited on the next line) the authors claim that Obp56f and CG11598 'were more expressed in D. melanogaster compared to D. simulans' but in the gel image the D. sim band is stronger for both genes (like D. sechellia) compared to the D. mel band. The authors also claim that the patterns of expression seen in the F1s are dominant for one allele and that this must be because of transvection. I agree this experiment is evidence for cis-regulatory change. However the interpretation that it is caused by transvection needs more explanation/justification and how do the authors rule out that it is not a cis X trans interaction between the species promoter differences and differences in the transcription factors of each species in the F1? Also my understanding is that transvection is relatively rare and yet the authors claim this is the explanation for 2/4 genes tested.
We appreciate the reviewer's comments on Figure 4 and the opportunity to improve its clarity. To address these concerns, we have carefully checked the figure citations and corrected any inconsistencies.
The reviewer raises an important point about our interpretation of transvection. We have expanded our discussion of this result to consider why transvection is a plausible explanation for the observed dominance patterns and also consider cis x trans interactions between species-specific promoters and transcription factor binding. While rare, transvection likely has more relevance in hybrid regulatory contexts involving homologous chromosome pairing which we discuss this in the revised text.
Line 112 states that the melanogaster subgroup contains 5 species - this is incorrect - while this study looked at 5 species there are more species in this subgroup such as mauritiana and santomea.
We have corrected the statement about the number of species in the melanogaster subgroup.
Lines 131-134 could explain better what the conservation scores and their groupings mean and the rationale for this approach.
We have clarified what the conservation scores and their groupings mean and the rationale for this approach.
Line 162 - the meaning of the sentence starting on this line is unclear - it sounds very circular.
We have rephrased the statement for more clarity.
Line 168 should cite Fig 4 H instead of F.
We have amended citation of Fig 4F to H.
Reviewer 3
In this study, McQuarrie et al. investigate the evolution of promoters of genes encoding accessory gland proteins (Acps) in species within the D. melanogaster subgroup. Using computational analyses and available genomic and transcriptomic datasets, they demonstrate that promoter regions of Acp genes are highly diverse compared to the promoters of other genes in the genome. They further show that this diversification correlates with changes in gene expression levels between closely related species. Complementing these computational analyses, the authors conduct experiments to test whether differences in expression levels of four Acp genes with highly diverged promoter regions are maintained in hybrids of closely related species. They find that while two Acp genes maintain their expression level differences in hybrids, the other two exhibit dominance of one allele. The authors attribute these findings to transvection. Based on their data, they conclude that rapid evolution of Acp gene promoters, rather than changes in trans, drives changes in Acp gene expression that contribute to speciation.
We thank Reviewer 3 for their thorough review and suggestions. We further thank the reviewer for acknowledging the importance of our findings and for pointing out that it contributes to our understanding of speciation. We have thoroughly addressed all comments from the reviewer and significantly revised the manuscript. We believe that this has greatly improved the manuscript.
Unfortunately, the presented data are not sufficient to fully support the conclusions. While many of the concerns can be addressed by revising the text to moderate the claims and acknowledge the methodological limitations, some key experiments require repetition with more controls, biological replicates, and statistical analyses to validate the findings.
Specifically, some of the main conclusions heavily rely on the RT-PCR experiments presented in Figure 4, which analyze the expression of four Acp genes in hybrid flies. The authors use PCR and RFLP to distinguish species-specific alleles but draw quantitative conclusions from what is essentially a qualitative experiment. There are several issues with this approach. First, the experiment includes only two biological replicates per sample, which is inadequate for robust statistical analysis. Second, the authors did not measure the intensity of the gel fragments, making it impossible to quantify allele-specific expression accurately. Third, no control genes were used as standards to ensure the comparability of samples.
The gold standard for quantifying allele-specific expression is using real-time PCR methods such as TaqMan assays, which allow precise SNP genotyping. To address this major limitation, the authors should ideally repeat the experiments using allele-specific real-time PCR assays. This would provide a reliable and quantitative measurement of allele-specific expression.
If the authors cannot implement real-time PCR, an alternative (though less rigorous) approach would be to continue using their current method with the following adjustments:
• Include a housekeeping gene in the analysis as an internal control (this would require identifying a region distinguishable by RFLP in the control).
• Quantify the intensity of the PCR products on the gel relative to the internal standard, ensuring proper normalization.
• Increase the sample size to allow for robust statistical analysis.
These experiments could be conducted relatively quickly and would significantly enhance the validity of the study's conclusions.
We thank the reviewer for their detailed suggestions for improving the conclusions in Fig. 4. Indeed, incorporating a housekeeping gene as a control supports our results for qualitative analysis of gene expression in hybrids assessing each allele individually (Fig 4), and improves interpretation for non-experts. We have also quantified differential gene expression in hybrids between species alleles and the log2 fold change from D. melanogaster. In addition, we have included an additional analysis in the new Fig. 5 which analyses RNA-seq expression changes in D. melanogaster x D. simulans hybrid male accessory glands. We believe these additions have significantly improved the manuscript and its conclusions.
While the following comments are not necessarily minor, they can be addressed through revisions to the text without requiring additional experimental work. Some comments are more conceptual in nature, while others concern the interpretation and presentation of the experimental results. They are provided in no particular order.
- A key limitation of this study is the use of RNA-seq datasets from whole adult flies for interspecies gene expression comparisons. Whole-body RNA-seq inherently averages gene expression across all tissues, potentially masking tissue-specific expression differences. While Acp genes are likely restricted to accessory glands, the non-Acp genes and the random gene sets used in the analysis may have broader expression profiles. As a result, their expression might be conserved in certain tissues while diverging in others- an aspect that whole-body RNA-seq cannot capture. The authors should acknowledge that tissue-specific RNA-seq analyses could provide a more precise understanding of expression divergence and potentially reveal reduced conservation when considering specific tissues independently.
We have added a section discussing the limitations in gene expression analysis in the discussion. In addition, we have included an additional Figure analysing gene expression in hybrid male accessory glands (Fig. 5).
- The statement in line 128, "Consistent with this model," does not accurately reflect the findings presented in Figures 2A and B. Specifically, the data in Figure 2A show that Acp gene expression divergence is significantly different from the divergence of non-Acp genes or a random sample only in the comparison between D. melanogaster and D. simulans. However, when these species are compared to D. yakuba, Acp gene expression divergence aligns with the divergence patterns of non-Acp genes or random samples. In contrast, Figure 2B shows that the distribution of expression changes is skewed for Acp genes compared to random control samples when D. melanogaster or D. simulans are compared to D. yakuba. However, this skew is absent when the two D. melanogaster and D. simulans are compared. Therefore, the statement in line 128 should be revised to accurately reflect these nuanced results and the trends shown in Figure 2A and B.
We have updated the statement for clarity. Here, the percentage of Acps showing significant gene expression changes is greater between more closely related species, but the distribution of expression changes increases between more distantly related species.
- The statement in lines 136-138, "Acps were enriched for significant expression changes in the faster evolving group across all species," while accurate, overlooks a key observation. This trend was also observed in other groups, including those with slower evolving promoters, in some of the species' comparisons. Therefore, the enrichment is not unique to Acps with rapidly evolving promoters, and this should be explicitly acknowledged in the text.
This is a valid point, and we have updated this statement as suggested.
- It would be helpful for the authors to explain the meaning of the d score at the beginning of the paragraph starting in line 131, to ensure clarity for readers unfamiliar with this metric.
This scoring method is described in the methods sections, and we have now included reference to thorough explanation of how d was calculated at the indicated section.
- In Figure 2C-E - the title of the Y-axis does not match the text. If it represents the percentage of genes with significant expression changes, as in Figure 2A, the discrepancies between the percentages in this figure and those in Figure 2A need to be addressed.
We have updated the method used to categorise significant changes in gene expression in the text and the figure legend for clarity.
- The experiment in Figure 3 needs a better explanation in the text. What is the analysis presented in Figure 3B-D. How many species were compared?
We have added additional details in the results section and an explanation of how sequence change hotspots were calculated in the results section is available.
- The concept of transvection should be omitted from this manuscript. First, the definition provided by the authors is inaccurate. Second, even if additional experiments were to convincingly show that one allele in hybrid animals is dominant over the other, there are alternative explanations for this phenomenon that do not involve transvection. The authors may propose transvection as a potential model in the discussion, but they should do so cautiously and explicitly acknowledge the possibility of other mechanisms.
We have updated the text to more conservatively discuss transvection, moving this to the discussion section with additional possibilities discussed.
- The statement at the end of the introduction is overly strong and would benefit from more cautious phrasing. For instance, it could be reworded as: "These findings suggest that promoter changes, rather than genomic background, play a significant role in driving expression changes, indicating that promoter evolution may contribute to the rise of new species."
We have reworded this line following the reviewer's suggestion.
- Line 32 of the abstract: The term "Acp" is introduced without explaining what it stands for. Please define it as "Accessory gland proteins (Acp)" when it first appears.
We have updated the manuscript to define Acp where it is first mentioned.
- Line 61: The phrase "...through relaxed,..." is unclear. Specify what is relaxed (e.g., "relaxed selective pressures").
We have included description of relaxed selective pressures.
- The sentence in lines 74-76, starting in "Interestingly,...." Needs revision for clarity.
We have removed the word interestingly.
- Line 112: Revise "we focused on the melanogaster subgroup which is made up of five species" to: "we focused on the melanogaster subgroup, which includes five species."
We have made this change in the text.
- In line 144 use the phrase "promoter conservation" instead of "promoter evolution"
We have updated the phrasing.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
In this study, McQuarrie et al. investigate the evolution of promoters of genes encoding accessory gland proteins (Acps) in species within the D. melanogaster subgroup. Using computational analyses and available genomic and transcriptomic datasets, they demonstrate that promoter regions of Acp genes are highly diverse compared to the promoters of other genes in the genome. They further show that this diversification correlates with changes in gene expression levels between closely related species. Complementing these computational analyses, the authors conduct experiments to test whether differences in expression levels of four Acp genes with highly diverged promoter regions are maintained in hybrids of closely related species. They find that while two Acp genes maintain their expression level differences in hybrids, the other two exhibit dominance of one allele. The authors attribute these findings to transvection. Based on their data, they conclude that rapid evolution of Acp gene promoters, rather than changes in trans, drives changes in Acp gene expression that contribute to speciation.
Major comments:
Unfortunately, the presented data are not sufficient to fully support the conclusions. While many of the concerns can be addressed by revising the text to moderate the claims and acknowledge the methodological limitations, some key experiments require repetition with more controls, biological replicates, and statistical analyses to validate the findings.
Specifically, some of the main conclusions heavily rely on the RT-PCR experiments presented in Figure 4, which analyze the expression of four Acp genes in hybrid flies. The authors use PCR and RFLP to distinguish species-specific alleles but draw quantitative conclusions from what is essentially a qualitative experiment. There are several issues with this approach. First, the experiment includes only two biological replicates per sample, which is inadequate for robust statistical analysis. Second, the authors did not measure the intensity of the gel fragments, making it impossible to quantify allele-specific expression accurately. Third, no control genes were used as standards to ensure the comparability of samples.
The gold standard for quantifying allele-specific expression is using real-time PCR methods such as TaqMan assays, which allow precise SNP genotyping. To address this major limitation, the authors should ideally repeat the experiments using allele-specific real-time PCR assays. This would provide a reliable and quantitative measurement of allele-specific expression.
If the authors cannot implement real-time PCR, an alternative (though less rigorous) approach would be to continue using their current method with the following adjustments:
Minor comments
While the following comments are not necessarily minor, they can be addressed through revisions to the text without requiring additional experimental work. Some comments are more conceptual in nature, while others concern the interpretation and presentation of the experimental results. They are provided in no particular order. 1. A key limitation of this study is the use of RNA-seq datasets from whole adult flies for interspecies gene expression comparisons. Whole-body RNA-seq inherently averages gene expression across all tissues, potentially masking tissue-specific expression differences. While Acp genes are likely restricted to accessory glands, the non-Acp genes and the random gene sets used in the analysis may have broader expression profiles. As a result, their expression might be conserved in certain tissues while diverging in others- an aspect that whole-body RNA-seq cannot capture. The authors should acknowledge that tissue-specific RNA-seq analyses could provide a more precise understanding of expression divergence and potentially reveal reduced conservation when considering specific tissues independently. 2. The statement in line 128, "Consistent with this model," does not accurately reflect the findings presented in Figures 2A and B. Specifically, the data in Figure 2A show that Acp gene expression divergence is significantly different from the divergence of non-Acp genes or a random sample only in the comparison between D. melanogaster and D. simulans. However, when these species are compared to D. yakuba, Acp gene expression divergence aligns with the divergence patterns of non-Acp genes or random samples. In contrast, Figure 2B shows that the distribution of expression changes is skewed for Acp genes compared to random control samples when D. melanogaster or D. simulans are compared to D. yakuba. However, this skew is absent when the two D. melanogaster and D. simulans are compared. Therefore, the statement in line 128 should be revised to accurately reflect these nuanced results and the trends shown in Figure 2A and B. 3. The statement in lines 136-138, "Acps were enriched for significant expression changes in the faster evolving group across all species," while accurate, overlooks a key observation. This trend was also observed in other groups, including those with slower evolving promoters, in some of the species' comparisons. Therefore, the enrichment is not unique to Acps with rapidly evolving promoters, and this should be explicitly acknowledged in the text. 4. It would be helpful for the authors to explain the meaning of the d score at the beginning of the paragraph starting in line 131, to ensure clarity for readers unfamiliar with this metric. 5. In Figure 2C-E - the title of the Y-axis does not match the text. If it represents the percentage of genes with significant expression changes, as in Figure 2A, the discrepancies between the percentages in this figure and those in Figure 2A need to be addressed. 6. The experiment in Figure 3 needs a better explanation in the text. What is the analysis presented in Figure 3B-D. How many species were compared? 7. The concept of transvection should be omitted from this manuscript. First, the definition provided by the authors is inaccurate. Second, even if additional experiments were to convincingly show that one allele in hybrid animals is dominant over the other, there are alternative explanations for this phenomenon that do not involve transvection. The authors may propose transvection as a potential model in the discussion, but they should do so cautiously and explicitly acknowledge the possibility of other mechanisms. 8. The statement at the end of the introduction is overly strong and would benefit from more cautious phrasing. For instance, it could be reworded as: "These findings suggest that promoter changes, rather than genomic background, play a significant role in driving expression changes, indicating that promoter evolution may contribute to the rise of new species."
Text edits:
Throughout the manuscripts there are incomplete sentences and sentences that are not clear. Below is a list of corrections:
This study addresses an important question in evolutionary biology: how seminal fluid proteins achieve rapid evolution despite showing limited adaptive changes in their coding regions. By focusing on accessory gland proteins (Acps) and examining their promoter regions, the authors suggest promoter-driven evolution as a potential mechanism for rapid seminal fluid protein diversification. While this hypothesis is intriguing and can contribute to our understanding of speciation, more rigorous analysis and experimental validation would be needed to support the conclusions. The revised manuscript can be of interest to fly geneticists and to scientists in the fields of gene regulation and evolution.
Keywords for my expertise: Enhancers, transcriptional regulation, development, evolution, Drosophila.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary
This manuscript explores promoter evolution of genes encoding seminal fluid proteins expressed in the male accessory gland of Drosophila and finds cis-regulatory changes underlie expression differences between species. Although these genes evolve rapidly it appears that the coding regions rarely show signs of positive selection inferring that changes in their expression and hence promoter sequences can underlie the evolution of their roles within and among species.
Major comments
Figure 1 illustrates evidence that the promoter regions of these gene have accumulated more changes than other sampled genes from the Drosophila genome. While this convinces that the region upstream of the transcription start site has diverged considerably in sequence (grey line compared to black line), Figure 1A also suggests the "Genespan" region which includes the 5'UTR but presumably also part of the coding region is also highly diverged. It would be useful to see how the pattern extends into the coding region further to compare further to the promoter region (although Fig 1H does illustrate this more convincingly).
Figure 2 presents evidence for significant changes in (presumably levels of) expression of male accessory gland protein (AcP) genes and ribosomal proteins genes between pairs of species, which is reflected in the skew of expression compared to randomly selected genes.
Figure 3 shows detailed analysis for 3 selected AcP genes with significantly diverged expression. The authors claim this shows 'substitution' hotspots in the promoter regions of all 3 genes but this could be better illustrated by extending the plots in B-D further upstream and downstream to compare to these regions.
Figure 4 shows the results of expression analysis in parental lines of each pair of species and F1 hybrids. However the results are very difficult to follow in the figure and in the relevant text. While the schemes in A, C. E and G are helpful, the gel images are not the best quality and interpretations confusing. An additional scheme is needed to illustrate hypothetical outcomes of trans change, cis change and transvection to help interpret the gels. On line 169 (presumably referring to panels D and F although C and D are cited on the next line) the authors claim that Obp56f and CG11598 'were more expressed in D. melanogaster compared to D. simulans' but in the gel image the D. sim band is stronger for both genes (like D. sechellia) compared to the D. mel band. The authors also claim that the patterns of expression seen in the F1s are dominant for one allele and that this must be because of transvection. I agree this experiment is evidence for cis-regulatory change. However the interpretation that it is caused by transvection needs more explanation/justification and how do the authors rule out that it is not a cis X trans interaction between the species promoter differences and differences in the transcription factors of each species in the F1? Also my understanding is that transvection is relatively rare and yet the authors claim this is the explanation for 2/4 genes tested.
Minor comments
Line 112 states that the melanogaster subgroup contains 5 species - this is incorrect - while this study looked at 5 species there are more species in this subgroup such as mauritiana and santomea.
Lines 131-134 could explain better what the conservation scores and their groupings mean and the rationale for this approach.
Line 162 - the meaning of the sentence starting on this line is unclear - it sounds very circular.
Line 168 should cite Fig 4 H instead of F.
This paper is generally well written although some sections would benefit from more explanation. The paper demonstrates cis-regulatory changes between the promoters of orthologs of male accessory gland genes underlie expression differences but that the species differences are not always reflected in hybrids, which the authors interpret as being caused by transvection although there could be other explanations. Overall this provides new insights into the regulation and divergence of these interesting genes. The paper does not explore the consequences of these changes in gene expression although this is discussed to some extent in the Discussion section.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The authors test the hypothesis that promoters of genes involved in insect accessory glands evolved more rapidly than other genes in the genome. They test this using a number of computational and experimental approaches, looking at different species within the Drosophila melanogaster complex. The authors find an increased amount of sequence divergence in promoters of accessory gland proteins. They show that the expression levels of these proteins are more variable among species than randomly selected proteins. Finally, they show that within interspecific hybrids, each copy of the gene maintains its species-specific expression level.
The work is done with expected standards of controls and analyses. The claims are supported by the analysis. My main criticism of the manuscript has to do not with the experiments or conclusion themselves but with the presentation. The manuscript is just not very well written, and following the logic of the arguments and results is challenging. The problem begins with the Abstract, which is representative of the general problems with the manuscript. The Abstract begins with general statements about the evolution of seminal fluid proteins, but then jumps to accessory glands and hybrids, without clarifying what taxon is being studied, and what hybrids they are talking about. Then, the acronym Acp is introduced without explanation. The last two sentences of the Abstract are very cumbersome and one has to reread them to understand how they link to the beginning of the Abstract.
More generally, if this reviewer is to be seen as an "average reader" of the paper, I really struggled through reading it, and did not understand many of the arguments or rationale until the second read-through, after I had already read the bottom line. The paragraph spanning lines 71-83 is another case in point. It is composed of a series of very strongly worded sentences, almost all starting with a modifier (unexpectedly, interestingly, moreover), and supported by citations, but the logical flow doesn't work. Again, reading the paragraph after I knew where the paper was going was clearer, but on a first read, it was just a list of disjointed statements.
Since most of the citations are from the authors' own work, I suspect they are assuming too much prior understanding on the part of the reader. I am sure that if the authors read through the manuscript again, trying to look through the eyes of an external reader, they will easily be able to improve the flow and readability of the text.
More specific comments:
The manuscript addresses a general question in evolutionary biology - do control regions diverge more quickly protein coding regions. The answer is that yes, they do, but this is actually not very surprising. The work is probably thus of more interest to people interested in the copulatory proteins or in the evolution of mating systems, than to people interested in broader evolutionary questions.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Below is a point-by-point response to reviewers concerns.
Main changes are colored in red in the revised manuscript.
Reviewer #1 (Significance (Required)):
General assessment:
This study provides a valuable computational framework for investigating the dynamic interplay between DNA replication and 3D genome architecture. While the current implementation focuses on Saccharomyces cerevisiae, whose genome organization differs significantly from mammalian systems.
Advance: providing the first in vivo experimental evidence in investigating the role(s) of Cohesin and Ctf4 in the coupling of sister replication forks.
Audience: broad interests; including DNA replication, 3D genome structure, and basic research
Expertise: DNA replication and DNA damage repair within the chromatin environment.
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
By developing a new genome-wide 3D polymer simulation framework, D'Asaro et al. investigated the spatiotemporal interplay between DNA replication and chromatin organization in budding yeast: (1) The simulations recapitulate fountain-like chromatin patterns around early replication origins, driven by colocalized sister replication forks. These findings align with Repli-HiC observations in human and mouse cells, yet the authors advance the field by demonstrating that these patterns are independent of Cohesin and Ctf4, underscoring replication itself as the primary driver. (2) Simulations reveal a replication "wave" where forks initially cluster near the spindle pole body (SPB) and redistribute during S-phase. While this spatial reorganization mirrors microscopy-derived replication foci (RFis), discrepancies in cluster sizes compared to super-resolution data suggest unresolved mechanistic nuances. (3) Replication transiently reduces chromatin mobility, attributed to sister chromatid intertwining rather than active forks.
This work bridges replication timing, 3D genome architecture, and chromatin dynamics, offering a quantitative framework to dissect replication-driven structural changes. This work provides additional insights into how replication shapes nuclear organization and vice versa, with implications for genome stability and regulation.
We thank Reviewer 1 for her/his enthusiasm and her/his comments that help us to greatly improve the manuscript.
However, the following revisions could strengthen the manuscript:
Major:
Generalizability to Other Species While the model successfully recapitulates yeast replication, its applicability to larger genomes (e.g., mammals) remains unclear. Testing the model against (Repli-HiC/ in situ HiC, and Repli-seq) data from other eukaryotes (particularly in mammalian cells) could enhance its broader relevance.
We agree with the reviewer that testing the model in higher eukaryotes would be highly informative. The availability of Repli-HiC on one hand and higher resolution microscopy on the other could enable insightful quantitative analyses. With our formalism, it is in principle already possible to capture realistic 1D replication dynamics as the integrated mathematical formalism (by Arbona et al. ref. [63]) was already used to model human genome S-phase. In addition, the formalism developed for chain duplication is generic and can be contextualized to any species. However, when addressing the problem in 3D, we would likely require including other crucial structural features such as TADs or compartments. Such a model would require an extensive characterization worthy of its own publication. These considerations are now mentioned in the Discussion as exciting future perspectives (Page 17).
On the other hand, we would like to highlight that, while very minimal in many aspects, our model includes many layers of complexity (explicit replication, different forks interactions, stochastic 1D replication dynamics, physical constraints at the nuclear level). In addition, addressing this problem in budding yeast offers the great advantage of simultaneously capturing at the same time both the local and global spatio-temporal properties of DNA replication and to focus first only on those aspects and not on the interplay with other mechanisms like A/B compartmentalization (absent in yeast) that may add confusions in the data analysis and comparison with experimental data . Studying such an interplay is a very important and challenging question that, we believe, goes beyond the scope of the present work.
Validation with Repli-HiC or Time-Resolved Techniques
The Hi-C data in early S-phase supports the model, but the intensity of replication-specific chromatin interactions is faint, which could be further validated using Repli-HiC, which captures interactions around replication forks. Alternatively, ChIA-PET or HiChIP targeting core component(s) (eg. PCNA or GINS) of replisomes may also solidify the coupling of sister replication forks.
We thank the reviewer for the suggestion. Unfortunately, corroborating our HiC results using Repli-HiC or HiChIP would require developing and adapting the protocols to budding yeast which is well beyond the scope of this work mainly focused on computational modelling. In addition, we believe that the signature found in our Hi-C data is clear and significant enough to demonstrate the effect.
However, we included in the Discussion (Page 15) a more detailed description on how our work compares with the Repli-HiC study in mammals. In particular, we added a new supplementary figure (new Fig. S23) where we discuss our prediction on how Repli-HiC maps would appear in yeast in both scenarios of sister-forks interaction. Interestingly, we find that:
1) Fountain signals are strongly enhanced when sister forks interact.
2) Only mild replication dependent enrichment is detected when diverging forks do not interact.
These two results imply that disrupting putative sister-forks interaction would have a drastic effect on Repli-HiC if compared to HiC.
Interactions Between Convergent Forks
The study focuses on sister-forks but overlooks convergent forks (forks moving toward each other from adjacent origins), whose coupling has been observed in Repli-HiC. Could the simulation detect the coupling of convergent fork dynamics?
We thank the reviewer for this suggestion. We included in our Hi-C analysis aggregate plots around termination sites. Interestingly, no clear signature of coupling between convergent forks was detected (such as type II fountains in mammals) in vivo and in silico. Similarly, from visual inspection of individual termination sites, no fountains were clearly observed. These results can be found in the new Fig. S24 and possible mechanistic explanations are described more in detail in the Discussion (Page 15).
Unexpected Increase in Fountain Intensity in Cohesin/Ctf4 Knockouts.
In Fig.3A, a schematic illustrating the cell treatment would improve clarity. In Sccl- and Ctf4-depleted cells, fountain signals persist or even intensify (Fig. 3A). This counterintuitive result warrants deeper investigation. Could the authors provide any suggestions or discussions? Potential explanations may include:
Compensatory mechanisms (e.g., other replisome proteins stabilizing sister-forks).
Altered chromatin mobility in mutants, enhancing Hi-C signal resolution.
Artifacts from incomplete depletion (western blots for Sccl/Ctf4 levels should be included).
A scheme illustrating the experimental protocol for degron systems (CDC45-miniAID & SCC1-V5-AID) with the corresponding western blots and cell-cycle progression are shown in Fig. S26. Note that for Ctf4, we are using a KO cell line where the gene was deleted.
We do agree with the reviewer that there exist several possible explanations explaining the differences between WT fountains and those observed in mutants. In the revised manuscript, we discussed some of them in Section 2 II B (Page 8):
(1) As already suggested in the paper, asynchronization of cells may impact the intensity of the fountains due a dilution effect mediated by the cells still in G1. Therefore, possible differences in the fractions of replicating/non-relicating cells between the different experiments (new Fig. S7C) would also result in differences in the signal. Moreover, it is important to highlight that aggregate plots are normalized (Observed/Expected) by the average signal (P(s)). Therefore, as Scc1-depleted cells do not exhibit cohesin-mediated loop-extrusion (see aggregate plots around CARs in new Fig. S7B), we may expect an enhancement of signal at origins due to dividing each pixel by a lower contact frequency with respect to the one found in WT.
(2) In the new Fig. S10, we plotted the relative enrichment of Hi-C reads around origins. While we already used the same approach to compare replicon sizes between simulations and experiments (see Fig S7A and response to comment n°9 of Reviewer 3), this analysis is instructive also when comparing different experimental conditions. While we find that the experiment in WT and Scc1-depleted cells show very similar replicon sizes, we do observe a small increase in the peak height for the cohesin mutant. This may also partially motivate differences in the intensity of the fountain. For ctf4Δ, we observe significantly smaller replicons. We speculate that such a mutant might exhibit slower replication and consequently might be enriched in sister-forks contacts.
(3) Compensatory mechanisms: we now briefly discussed this in the Discussion (Page 15).
Inconsistent Figure References
Several figure citations are mismatched. For instance, Fig. S1A has not been cited in the manuscript. Moreover, there is no Fig.1E in figure 1, while it has been cited in the text. All figure/panel references must be cross-checked and corrected.
We thank the reviewer for this observation. We have now corrected the mismatches.
Minor:
Page2: "While G1 chromosomes lack of structural features such as TADs or loops [3]" However, Micro-C captures chromatin loops, although much smaller than those in mammalian cells, within budding yeast.
Loops of approx 20-40 kb are found in interphase in budding yeast but only after the onset of S-phase ( ref. [52-61]). For this reason, our G1 model of yeast without loops well captures the experimental P(s) curves (Fig. S2). See also answer to point 12 of reviewer 2 .
In figure 2E, chromatin fountain signals can be readily observed in the fork coupling situation and movement can also be observed. However, the authors should indicate the location of DNA replication termination sites and show some examples at certain loci but not only the aggregated analysis.
The initial use of aggregate plots was motivated by the fact that fountains are quite difficult to observe at the single origin level in the experimental Hi-C due to the strong intensity of surrounding contacts (along the diagonal). However, when dividing early-S phase maps by the corresponding G1 map, we can now observe clear correlation between origin and fountain positions on such normalized maps. We now added an example for chromosome 7 in Fig.3 indicating early/late origins.
In Fig. S8 and S9 (where we also included termination sites), we show that fountains are prominently found at origins during S-phase and are lost in G2/M.
Reviewer #2 (Significance (Required)):
The topic is relevant and the problem being addressed is very interesting. While there has been some earlier work in this area, the polymer simulation approach used here is novel. The simulation methodology is technically sound and appropriate for the problem. Results are novel. The authors compare their simulations with experimental data and explore both interacting and non-interacting replication forks. Most conclusions are supported by the data presented. Reviewer #2 (Evidence, reproducibility and clarity (Required)):
The manuscript by D'Asaro et al. investigates the relationship between DNA replication and chromatin organization using polymer simulations. While this is primarily a simulation-based study, the authors also present relevant comparisons with experimental data and explore mechanistic aspects of replication fork interactions.
We thank Reviewer 2 for her/his positive evaluation of our work and her/his suggestions that help us to clarify many aspects in our manuscript.
The primary weakness is that many aspects are not clear from the manuscript. Below is a list of questions that the authors must clarify:
In the Model and Methods section, it is written "Arbitrarily, we choose the backbone to be divided into two equally long arms, in random directions." It is unclear what is meant by "backbone to be divided" and "two equally long arms." Does this refer to replication?
We agree with the reviewer that the term backbone may be ambiguous. In the context of the initialization of the polymer, it refers to the L/4 initial bonds used to recursively build an unknotted polymer chain of final size L using the Hedgehog algorithm (see refs [101,109]). As shown in the Fig S1A, these initial L/4 bonds define the initial backbone of each chromosome before they are recursively grown to their final size. We chose to divide them into two branches (called “arms” in the old version of the manuscript) of equal length (L/8) and with random orientations. To avoid any ambiguity between the term arm used in that context and the chromosome arms in a biological sense (sequences on the left and right with respect to centromeres), we changed it to “linear branches” to improve clarity. We highlighted in Fig. S1A two examples of such a “V-shaped” backbone.
As stated in the text, these initial configurations are artificial and just aim to generate unknotted, random structures. After initiating the structures, we then added the geometrical constraints to the centromeric, telomeric and rDNA beads. This, combined with the tendency of the polymer to explore and fill the spherical volume, determine the relaxed G1-like state (see Fig. S2) obtained after an equilibration stage (corresponding to 10^7 MCS). Only after that initialization protocol, DNA replication is activated.
In chromosome 12, since the length inside the nucleolus (rDNA) is finite, the entry and exit points should be constrained. Have the authors applied any relevant constraint in the model?
Indeed, we did not introduce any specific constraint on the relative distance between rDNA boundary monomers in our model. They can therefore freely diffuse, independently from each other, on the nucleolus surface. This point is now clarified in the text. Note that, in this paper, we did not aim to finely describe the rDNA organization and its interactions with the rest of the genome, that is why we did not explicitly model rDNA. Moreover, to the best of our knowledge, there is not available experimental data to potentially tune such additional restraints.
Previous models such as Tjong et al. (ref. [66]) and Di Stefano et al. (ref [67]) have used very similar approximations than us. In the works of Wong et al. (ref.[61]) and Arbona et al. (ref.[63]), rDNA is explicitly modelled via larger/thicker beads/segments, and thus accounts for some generic polymer-based constraints between rDNA boundary elements.
However, note that all these different models, including ours, still correctly predict the strong depletion of contacts between rDNA boundaries, indicating that there exists a spatial separation between the two boundary elements that is qualitatively well captured by our model (See Fig. S1 D and Fig. 1B).
What is the rationale for normalizing the experimental and simulation results by dividing by the respective P_intra(s = 10 kb)?
This normalization was used in Fig. 1 to obtain a rescaling between experiments and simulations. This approach assumes that simulated and experimental Hi-C maps are proportional by a factor that, in Fig 1B, was set to P_exp(s=16kb)/P_sim(s=16kb). Similar strategies are used in a number of modeling studies (for example ref. [103,106]).
We use the average contact frequency (P_intra) at this genomic scale (s in the order of 10s of kb) because our polymer simulations well capture the experimental P(s) decay above this scale. This method allows to plot the two signals with the same color scale and to give a qualitative, visual intuition on the quality of the modeling. Note that normalization has no impact on the Pearson correlation given in text. More generally, it allows to semi-quantitatively compare predicted and experimental Hi-C data.
In Fig 1D, we instead normalize the average signal between pairs of centromeres (inter-chromosomal aggregate plot off-diagonal) by the average P_intra(s=10kb). This method allows estimating how frequently centromeres of different chromosomes are in contact relative to intra-chromosomal contacts at the chosen scale (10 kb). In the new paragraph “Comparison with in vivo HiC maps in G1” (Page 22) , we describe more in detail the quantitative insights that can be recovered from such analysis.
As a comparison, such normalization is not required when computing Observed/Expected maps (Fig. 1C or aggregate plots in Fig. 2 and Fig. 3) as simulation and experimental maps are normalized by their own P(s) curves. We now clarify this aspect in the Materials in Methods under the paragraph “Comparison between on diagonal aggregate plots” (Page 22).
In the sentence "For instance, chromosomes are strictly bound by the strong potential to localize between 250 and 320 nm from the SPB," is it 320 or 325 nm? Is there a typo?
We confirm that the upper bound is indeed 325 nm as stated in Eq.2 and not 320 nm.
Please list the number of beads in each chromosome and the location of the centromere beads.
A new table (Table S2) was included to highlight beads number and centromere positions.
In Eq. 7, when the Euclidean distance between the sister forks d_ij > 50 nm, the energy becomes more and more negative. This implies that the preferred state of sister forks is at distances much greater than 50 nm. Then how is "co-localization of sister forks" maintained?
We corrected the typo sign in Eq.7. The corrected equation without the minus sign - consistently with what simulated - implies that sister forks tend to minimize their 3D distance. The term goes to zero when their distance is within 40 nm (2 nearest-neighbouring sites).
The section on "non-specific fork interactions" is unclear. You state that the interaction is between "all the replication forks in the system," but f_ij is non-zero only for second nearest-neighbors. The whole subsection needs clarification.
We corrected the text, specifying that the energy is non-zero for both first and second neighbours. In practice, two given forks do not experience any attractive energy unless their 3D distance is less than 2 nearest-neighbours. To clarify this aspect, we articulated more in the methods how non-specific fork interactions are implemented in the lattice during the KMC algorithm. We also included a new supplementary image (Fig. S15), where we schematize how forks move in 3D and how changes in their position update the table that tracks the number of forks around each lattice site.
Eq. 6 has no H_{sister-forks}. Is this a typo?
We confirm that it is a typo and the formula was corrected to H_{sister-forks}.
While discussing the published work, the authors may cite the recent paper [https://doi.org/10.1103/PhysRevE.111.054413].
The reference is now included when discussing previous polymer models of DNA replication.
It is not clear how the authors actually increase the length of new DNA in a time-dependent manner. For example, when a new monomer is added near the replication origin (green bead in Fig. 3C), what happens to the red and blue polymer segments? Do they get shifted? How do the authors take into account self-avoidance while adding a new monomer? These details are not clear.
The detailed description of the chain duplication algorithm and its systematic analysis was performed in our previous study (ref. [25]).
However, we agree with the reviewer that to improve self-consistency more details must be included in the present manuscript (see also answer to comment 1 of Reviewer 3). In particular, we now highlight in Materials and Methods that self-avoidance is indeed temporarily broken when we add a newly replicated monomer on top of the site where the fork is. Such double occupancy in the lattice rapidly vanishes due to 3D local moves. We refer to our PRX work (ref [25] and in particular to the following figure (extracted from FIG. S1 in ref.[25]) which illustrates how the bonds/segments of the two sister chromatids are consistently maintained.
How do the authors ensure that monomers get added at a rate corresponding to velocity v? The manuscript mentions "1 MCS = 0.075 msec," but in how many MC steps is a new monomer added? How is it decided?
Similarly to origin firing, replication by fork movement along the genome occurs stochastically, with a rate which we derive by converting the physiological fork speed in yeast 2.2 kb/min (ref. [41]) into a rate in (number of monomer/MCS) units. In practice, we generate a random number that, if smaller than such a rate, leads to forks duplication. We clarify this aspect in the Materials and Methods, also referring to our previous work for a more detailed summary.
The authors stress the relevance of loop extrusion. However, in their polymer simulation, the newly replicated chromatin does not form any loops. Is this consistent with what is known?
Indeed, our simulations do not have any concurrent extrusion mechanism such as cohesin-mediated loops. This choice was purposely made to isolate and characterize replication-dependent effects.
That is why we compare our predictions on chromatin fountain patterns (Fig. 3) with data obtained for the Scc1 mutant strain where cohesin is absent in order to disentangle the possible interference with loop-extruding cohesin. For subsection C where microscopy data are available only in WT condition, we cannot rule out that the observed discrepancies between experiments and predictions cannot be due to missing mechanisms including loop extrusion. It was already mentioned in the Discussion (Page 16). It is however unclear whether sparse and small loops between CARs (see Fig. S7B) in S-phase, could be sufficient to recapitulate the microscopy estimates on the sizes of replication foci and no clear signature of inter-origin loops (possibly mediated by loop extrusion) are observed in Hi-C data in WT and Scc1 deficient conditions.
Moreover, as mentioned in the Discussion, the poorly characterized mechanisms behind forks/extruding-cohesin encounters does not allow for a straightforward modelling of such processes whose accurate description/simulation would require its own study.
Please add a color bar to Fig. 4B.
The color bar was included.
In the MSD plot (Fig. 6), even though it appears to be a log-log plot, the exponents are not computed. Typically, exponents define the dynamics.
We plot the expected 0.5 exponent at smaller time-scales as mentioned in the main text in Fig. 6, previously included only in new Fig. S19A.
The dynamics will depend on the precise nature of interactions, such as the presence or absence of loop extrusion. If the authors present dynamics without extrusion, is it likely to be correct?
The reviewer is correct in highlighting how our model does not capture the potential decrease in dynamics due to cohesin mediated loop extrusion. However, our model does capture the expected Rouse regime (see Fig. 6A, S19A and ref [83]), which justify our timemapping strategy. In comment 16 of reviewer 3, we discuss more in detail the robustness of our results with respect to variation in such a mapping. In the specific context of Fig. 6A, we predict the gradual decrease in dynamics due to sister chromatids intertwining independently of any cohesin-associated activity (both loop-extruding and cohesive). As loop extrusion is also decreasing chromatin mobility overall (ref. [87]), if such a decrease in mobility is observed in WT in vivo, it may be indeed difficult to assign such a decrease to replication rather than loop extrusion. That is why in the Discussion (Page 16), we propose to compare our prediction to experiments in cohesin-depleted cells. In the context of Fig.6B&C, we don’t expect loop extrusion to be a confounding effect as the predicted decrease in dynamics is specific to forks.
Reviewer #3 (Significance (Required)):
The work has been conducted thoroughly, and in general the paper is well written with good attention to detail. As far as I am aware, this is the first study where replication is simulated in a whole nucleus context, and the scale of the simulations is impressive. This allows the authors to address questions on replication foci and the spatiotemporal organisation of replication which would not be possible with more limited simulations, and to compare the model with previous experimental work. This, together with the new HiC data, I think this makes this a strong paper which will be of interest to biophysics and molecular biology researchers; the manuscript is written such that it would suit an interdisciplinary basic research audience.
We thank Reviewer 3 for her/his enthusiasm and her/his comments that help us to greatly improve the manuscript.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
The paper "Genome-wide modelling of DNA replication in space and time confirms the emergence of replication specific patterns in vivo in eukaryotes" by D'Asaro et. al presents new computational and experimental results on the dynamics of genome replication in yeast. The authors present whole-nucleus scale simulations using a kinetic Monte Carlo polymer physics model. New HiC data for synchronised yeast samples with different protein knock-downs are also presented.
The main questions which the paper addresses are whether sister forks remain associated during replication, whether there is more general clustering of replication forks, and whether replication occurs in a 'spatial wave' through the nucleus. While the authors' model data are not able to conclusively show whether sister forks remain co-localised, the work provides some important insights which will be of high interest to the field.
I have no major issues with the paper, only some minor comments and suggestions to improve the readability of the manuscript or provide additional detail which will be of interest to readers. I list these here in the order in which they appear in the paper. There are also a number of typos and grammatical issues through the text, so I recommend thorough proofreading.
The paper seems to be aimed at a broad interdisciplinary audience of biophysicists and molecular biologists. For this reason, the introduction could be expanded slightly to include some more background on DNA replication, the key players and terminology. Also, it seems that this work builds on previous modelling work (Ref. 19), so a bit more detail of what was done there, and what is new here would be helpful. The final paragraph the introduction mentions chromosome features such as TADs and loops, which should be explained in more detail.
We now have expanded the introduction to address some of these aspects. In particular, also as a response to comment 1 of Reviewer 4, we included additional background on the eukaryotic replication time program. We address in more detail its known interplay and correlation with crucial 3D structural features such as compartments and TADs. Finally, we add a sentence to clarify how the current work is distinct from the prior implementation and the novelty introduced here.
In the first results section, end of p2, the "typical brush-like architecture" is mentioned. This is not well explained, some additional detail or a diagram might help.
As very briefly summarized in the mentioned paragraph, the yeast genome is organized in the so-called Rabl organization where chromosome arms are all connected via the centromeres at the Spindle Pole Body (SPB). This is analogous to the definition of a polymer brush where several branches (the arms in this case), are grafted to a surface or to another polymer (see new Inset panel in Fig S1B). We refer in the main text to the scheme in Fig. S1B where we also include the snapshot of a single chromosome and the physical constraints that characterize this large-scale organization and extend the caption to clarify the analogy. A typical emerging feature at the single chromosome level is described in Fig. 1 B and C.
On p3-4, some previous work is described, with Pearson correlations of 0.86 and 0.94 are mentioned. What cases these two different values correspond to is not clear.
These Pearson correlations are obtained for our own modeling. We correct the values in the main text and more clearly indicate the specific correspondence with the maps used. We describe now in the Materials and Methods (new paragraph “Comparison with in vivo HiC maps in G1” and Table S2) how these values were obtained.
In section II-A-2, on the modelling details, it should be made clearer that the nucleus volume is kept constant, and that this is an approximation since typically the nucleus grows during S-phase. This is discussed in the Methods section, but it would be useful to also mention it here (and give some justification why it will not likely change the results).
We now state more clearly in the main text the limitation of our model regarding the doubling of DNA content without any increase of nuclear size. As mentioned in the Discussion, we do not expect this approximation to strongly impact our results, which mainly focus on early S-phase.
We now also included in the Discussion how the detection of the “replication wave” should be qualitatively independent of the density regime. In fact, even in the case of growing nuclei and constant density, the polarity induced by the Rabl organization and replication timing are the main drivers of such fork redistribution.
Regarding the slowdowning in diffusion due to sister chromatids intertwinings (see response to comment 13), we instead verified that the effect is indeed density independent (new Fig S21).
Fig 2. The text in Fig 2B is much smaller than other panels and difficult to read. Also Fig 3B, Fig 6.
This is now corrected.
In 2E, are the times given above each map the range which is averaged over? This could be clearer in the caption. In the caption it stated that these are 'observed over expected'; what the 'expected' is could be clearer.
We reformulate the description in the caption to make clearer that the time indicated above the plots indicate the time window used for the computation. As mentioned more in detail in the response to comment 17 below (and comment 3 of Reviewer 2), we included in the Material and Methods a more precise description on the normalization used in the case of on-diagonal aggregate plots (observed-over-expected).
In section II-B-2, the authors state that the cells are fixed 20 mins after release from S-phase. Can they comment on the rationale behind this choice, since from Fig 2 their simulations predict that the fountain pattern will no-longer be visible by that time.
In the experimental setup, cells are arrested in G1 with alpha-factor and then released in S-phase (see Fig S26 with corresponding scheme). The release from G1 synchronisation is not immediate, and staging of cells by flow-cytometry every 5 minutes for 30 minutes after release (data not shown in the main text but provided below) proved 20 minutes to be an adequate early S-phase timepoint (Page 17 in the Materials and Methods). As a consequence, the times indicated when describing the in vivo experiment, do not correspond to the ones indicated in our in silico system, for which the onset of replication is well defined. For these reasons, we have to determine which time window among the ones used in Fig 2E, is the most appropriate to compare with the experiment (see response to comment 9 for more details).
Fig.R1: Cell cycle progression monitored by flow cytometry after the release. For the first 15 minutes, cells are still mainly in G1 and only start replicating ~20 minutes after the release.
Section II-B-2(b) could be clearer. I don't understand what the conclusion the authors take from the metaphase arrest maps is. I'm not sure why they discuss again the Cdc45-depleted cells here, since this was already covered in the previous section.
Taken together, the G1, Cdc20 (metaphase-arrested cells), and Cdc45-depleted (early S cells but not replicated) conditions suggest that fountains reflect ongoing replication. Namely, G1-arrest shows that fountains require S-phase entry; Cdc45-depletion shows that fountains require origin firing and is not due to another S-phase event; and metaphase-arrested cells show that fountains are not permanent structures established by replication, but a transient replication-dependent structure.
This demonstrates that the emerging signal is not trivially dependent on (1) the presence of the second sister chromatids; or on (2) potential overlaps between origin positions and barriers (CARs) to loop extrusion (see also comment 12 of Reviewer 2). A sentence at the end of II-a was added to clarify the different information gained with the two strains.
We discuss again the cdc20 and cdc45 mutants in II-b to highlight how the results in II-a do not exclude potential interplay between cohesin-mediated loop-extrusion in presence forks progression. These considerations motivated our experiment in Scc1-depleted cells during early S-phase.
At the start of p8 (II-B-3) there is a discussion of the mapping to times to the early-S stage experiments. This could have more explanation. I don't follow what the issue is, or the process which has been used to do the mapping. From Fig 2B, it seems that the simulation time is already mapped well to real time.
As mentioned above in comment 7, we cannot clearly define a “t=0” when replication starts in vivo as the release from the G1-arrest is not immediate and perfectly synchronous. On the other hand, the times indicated within the text are those following the onset of polymer self-duplication in our simulations. Note that the mean replication time (MRT) shown in Fig.2B does not represent an absolute time, but rather an average relative timing along S-phase (signal rescaled between 0 and 1).
For all these considerations, we think that the most reliable strategy to compare fountains in vivo and in silico is to look at the replicon size via the enrichment in raw contacts around early origins, as illustrated in Fig S7A. In practice, looking at the relative counts of contacts around early origins we have a proxy for the average replicon size that we can match by computing the same analysis on simulated signals (Fig S7A). As a result, we find that the best simulated time window is between 5 and 7.5 minutes, compatible with early-S phase and with an approximate duration of G1 after release of 15 minutes as observed in other studies (ref. [61]).
Note that our conclusions are robust with respect to modulating this mapping method. In particular in Fig. S7, we thoroughly investigated how several confounding factors (such as time window used or partial synchronization) may impact the quantitative nature of our prediction without affecting the qualitative insights.
We included a more precise reference to the Supplementary Materials, where the approach is described and clarified.
In Fig 4A above each plot there is a cartoon showing the fork scenario. The left-hand cartoon is rendered properly, but the right-hand one has overlapping black boxes which I don't think should be there. These black boxes are present in many other figures (4B, 3B, 2E etc).
This issue seems to appear using the default PDF viewer on Mac OS. We have corrected the problem and no more black boxes should appear in the main text and in the Supplementary Material.
In II-C-2(b) it is mentioned that the number of forks within RFis is always assumed to be even. This discussion could be clearer. In particular, the authors state that under both fork scenarios, in the simulations they can detect odd numbers of forks within RFis - how can this happen in the case where sister forks are held together?
We included a more accurate description in the main text about why Saner et al. (ref [20]) make these assumptions in their estimates. We highlight possible inconsistencies such as the presence of termination events which, in our formalism, break sister forks interactions and lead to single forks to be detected. We also clarify the latter point when describing Fig 5B and describe in more detail replication bubbles merging events in the Materials and Methods.
Fig 6B and C, it would be useful if the same scale was used on both plots.
We now use the same scale when plotting Fig 6B and C.
Section II-D-1. There is a discussion on the presence of catenated chains; I did not understand how the replicated DNA becomes catenated, and what this actually means in this context. The way the process is described and the snapshots in Fig2C do not suggest that the chains are catenated. Some further discussion or a diagram would be useful here.
We included a small paragraph to better explain how intertwining of sister chromatids occurs, and more clearly refer to a snapshot in supplementary figure S19D (Page 14). As correctly mentioned by the reviewer, replication bubbles by construction are always unknotted during their growth (see example in Fig. 2C). As we thoroughly characterize in our previous work (ref. [25]), when several replication bubbles merge, the random orientation of sister chromatids potentially lead to catenation points and intertwined structures. We show below a scheme from our previous work (ref [25]). While in this past work, we demonstrated that the center of mass of the two sister chromatids show subdiffusive behaviour due to the additional topological constraints of their intertwining, this new analysis in the present work suggests that possible effects may also be observed when tracking the MSD (mean square displacement at the locus level) in a more realistic scenario where we included correct replication timing, chromosome sizes and Rabl-organization.
On p14 (section III) there is a section discussing possible mechanisms for sister fork interactions, and that result that Ctf4 might not play a role in this, as previously suggested. Are there any other candidate proteins which could be tested in the future?
To the best of our knowledge, there is no other candidate protein of the replisome that has been directly associated to sister-fork pairing in previous studies (as Ctf4). However, components of the replisome such as Cdt1, that have the capacity to oligomerize/self-interact, could be good candidates. We now mention this possibility in the Discussion (Page 15).
As on p14, second paragraph: there is a sentence "replication wave [51] cannot be easily visualised at the single cell level.", which seems to contradict the discussion on p9 "such a "wave" can also be observed at the level of an individual trajectory (Video S3,4) even if much more stochastic." I think more explanation is needed here.
We rephrased the mentioned passages to clarify the differences in detecting such “replication wave” at the population vs single cell level. In video S3 and S4, we can still observe an enrichment of forks at the SPB and later in S-phase a shift towards the equatorial plane. However, the stochasticity of polymer dynamics and 1D replication strongly hinder the ability to clearly visualize such redistribution.
In the methods section, p18, it is mentioned that the volume fraction is 3%. I assume this is before replication, and so after replication is complete this will increase to 6%. This should be stated more explicitly, with also a comment on the 5% volume fraction used in the time-scale mapping discussed on p17.
Indeed, we choose to map the experimental MSD measured in ref [83] by simulating a homopolymer 5% volume fraction and in periodic boundary conditions for consistency to previous work in the group (ref. [102-106]) and our previous replication model (ref.[25]). Moreover, this intermediate density regime also lies in between the minimal (3%) and maximal (6%) densities present in our system. When redoing the time mapping with the G1 MSD plotted in Fig 6A and new Fig S19A, we obtain a very similar value of approx. 1MC=0.6ms. Note that the time mapping aims to obtain a rough estimation of real times as several factors, such as active processes, non-constant density, cell-cycle progression may all contribute to chromatin diffusion in vivo (see also comment 15 to Reviewer 2). In the context of our formalism, differences in time mapping do not affect the 1D replication dynamics as all the parameters to model the 1D process are rescaled by the same factor. Moreover, as we characterized in more depth in our previous work (ref [25]), a crucial aspect that defines self-replicating polymers is the relationship between fork progression and the polymer relaxation dynamics. In physiological conditions, we remain in the regime where forks progress almost quasi-statically to allow the bubbles to re-equilibrate. Therefore, small discrepancies in the time mapping will not modify this regime and our results should remain robust.
On p20, processing of simulated HiC using cooltools is discussed. For readers unfamiliar with this software, a bit more detail should be given. Specifically, how does the normalisation account for having some segments which have been replicated and some which have not. Later on the same page (IV-C-2) two different strategies for comparing HiC maps are given; why are two different methods required, and what is the reasoning in each case?
In the raw - unbalanced - data, we observe an artificial increase in contacts around origins in S-phase for both simulation and experiments. This is simply due to the presence of the second Sister chromatids and the fact that contacts between distinct DNA segments are mapped to a single bin.
In the new Fig. S25, we illustrate this effect by computing aggregate plots around early origins using single-chromosome simulations. We demonstrate that the ICE normalization corrects for the variations in copy number due to replication and thus for such artificial increases in contacts during S-phase. We show that such a normalization is equivalent to explicitly divide each bin by the average copy-number of the corresponding segments.
We have now included a sentence in the Materials and Methods to clarify this. Moreover, a detailed description of the other alternative strategies used to compare experiments and simulations were presented in response to comment 3 to Reviewer 2 and two new paragraphs were added in the Materials and Methods.
The references section has an unusual formatting with journal names underlined.
We updated the formatting.
Reviewer #4 (Significance (Required)):
D’Asaro et al focus on the problem of how genome structure is altered by the progression of replisomes through S-phase in the budding yeast S. cerevisiae. The authors employ computational polymer modeling of G1 chromosomes, then implement a hierarchical model of replication origin firing along these polymers to examine how the G1 chromosome structural state is perturbed by replisome progression. Their results indicate that replication origins create 'fountains' - Hi-C map features that other groups have demonstrated are likely to originate from symmetric extrusion by condensin / cohesin complexes originating at a fixed point. These 'fountains' appear to be cohesin-independent, as revealed by depletion Hi-C experiments. Finally, the authors provide evidence from their model of a 'replication wave' that emanates from the spindle pole body. This is an interesting manuscript that raises some exciting questions for the field to follow up on.
Reviewer #4 (Evidence, reproducibility and clarity (Required)):
In their manuscript, "Genome-wide modeling of DNA replication in space and time confirms the emergence of replication specific patterns in vivo in eukaryotes," authors Asaro et al perform computational modeling analyses to address an important open question in the chromatin field: how is DNA replication timing coupled to 3D genome architecture? Over the past ten years, the convergence of high-resolution replication timing (RT) analysis with high-resolution 3D genome mapping (e.g. 'Hi-C' technology) has resulted in the discovery that replication timing domains overlap considerably with 3D genomic domains such as topologically associating domains (TADs). How and why this happens both remain unknown, and advances in 3D genome mapping technology have provided even more data to model the problem of both 1) scheduling replication from distinct series of origins / initiation zones, and 2) modeling how 3D genome architecture is altered by the progression of replication forks, which inherently destroy chromatin structure before faithfully reforming G1 structures on daughter chromatids. As such, the problem being tackled by this computational manuscript is interesting.
We thank Reviewer 4 for her/his positive evaluation of our work and her/his comments that help us to greatly improve the manuscript.
Reviewer Comments / Significance
In their manuscript, "Genome-wide modeling of DNA replication in space and time confirms the emergence of replication specific patterns in vivo in eukaryotes," authors D’Asaro et al perform computational modeling analyses to address an important open question in the chromatin field: how is DNA replication timing coupled to 3D genome architecture? Over the past ten years, the convergence of high-resolution replication timing (RT) analysis with high-resolution 3D genome mapping (e.g. 'Hi-C' technology) has resulted in the discovery that replication timing domains overlap considerably with 3D genomic domains such as topologically associating domains (TADs). How and why this happens both remain unknown, and advances in 3D genome mapping technology have provided even more data to model the problem of both 1) scheduling replication from distinct series of origins / initiation zones, and 2) modeling how 3D genome architecture is altered by the progression of replication forks, which inherently destroy chromatin structure before faithfully reforming G1 structures on daughter chromatids. As such, the problem being tackled by this computational manuscript is interesting.
D’Asaro et al focus on the problem of how genome structure is altered by the progression of replisomes through S-phase in the budding yeast S. cerevisiae. The authors employ computational polymer modeling of G1 chromosomes, then implement a hierarchical model of replication origin firing along these polymers to examine how the G1 chromosome structural state is perturbed by replisome progression. Their results indicate that replication origins create 'fountains' - Hi-C map features that other groups have demonstrated are likely to originate from symmetric extrusion by condesin / cohesin complexes originating at a fixed point. These 'fountains' appear to be cohesin-independent, as revealed by depletion Hi-C experiments. Finally, the authors provide evidence from their model of a 'replication wave' that emanates from the spindle pole body. This is an interesting manuscript that raises some exciting questions for the field to follow up on.
Major Comments
There is a tremendous amount of work coupling RT domains to 3D genome architecture, especially deriving from the ENCODE and 4D Nucleome consortia. These studies are not adequately highlighted in the introduction and discussion of this manuscript, and this treatment of the literature would ideally be amended in any revised manuscript.
We include new sentences in the introduction to discuss more in detail the correlation between 3D genome architecture and replication timing program, and advancement in this field in the last decades. We also included additional citations to reviews and publications (ref [8-16]). These references were also included at the end of the Discussion where we address the exciting perspective of employing our model in higher eukaryotes and potentially tackle the complex interplay between 3D nuclear compartmentalization and replication dynamics (see also response 1 to Reviewer 1).
S. cerevisiae origins of replication differ from metazoan origins of replication in that they are sequence-defined and are known to fire in a largely deterministic pattern (see classic study PMID11588253). From the methods of the authors it is not clear that the known deterministic firing pattern is being used here, but instead a stochastic sampling method? Please clarify in the manuscript. Specifically, it would be good to understand how the Initiation Probability Landscape Signal correlates with what is already known about origin firing timing.
In our model, the positions of origins are stochastically sampled proportionally to the IPLS which was inferred directly from experimental MRT (ref. [63]) and RFD (ref. [44]). This modeling approach allows reproducing with a very high accuracy the known replication timing data (correlation of 0.96) and Fork directionality data (correlation of 0.91) (see ref. [71]). Origins were defined as the peaks in the IPLS signal. In Fig S3, we extensively compare these origins and the known ARS positions from the Oridb database. For example, most of our early origins (96%) are located close to known, confirmed ARS. Moreover, even if our algorithm is stochastic for origin firing, we remark that each early origin will fire in 90 % of the simulations, coherent with the quasi-deterministic pattern of origin firing and experimental MRT and RFD data. We now have added such statistics of firing in the revised manuscript (Page 4).
It seems possible that experimental sister chromatid Hi-C data (PMID32968250) and nanopore replicon data (PMID35240057) could be used to further ascertain the validity of some of the findings of this paper. Specifically, could the authors demonstrate evidence in sister chromatid Hi-C data that the replisome is in fact extruding sister chromatids? Moreover, are the interactions being measured specifically in cis (as opposed to trans sister contacts)? For the nanopore replicon data, how do replicon length, replication timing, and position along the replication 'wave' correlate?
We thank the reviewer for the suggestions.
Hopelessly there is currently no Sister-C data available during S-phase. In the seminal study (PMID32968250), cells were arrested in G2/M via nocodazole treatment. For a different unpublished work, we already analysed in detail the SisterC dataset and we did not observe clear fountain-like signature, consistent with our own G2/M Hi-C maps (cdc20) where fountains were absent. Note that, in the present work, in order to compare our predictions with standard HiC data, we included all contacts (cis and trans chromatids), mapping pairwise contacts from distinct replicated sequences/monomers to a single bin (see also response to comment 17 to Reviewer 3 and new Fig. S25).
We now mention in the Discussion that Sister-C data during S-phase could help monitoring the role of replisomes on relative sister-chromatids organization (Page 15).
Main results from the nanopore replicon data study include the observed high symmetry between sister forks and their linear progression, as the density of replicons appears to be uniform with respect to their length. Since these two specific constraints are already present in the framework of Arbona et al. (ref. [63]), our model is able to reproduce these features of DNA replication captured by the nanopore data.
Moreover, as we model with very high accuracy replication timing data (see response to comment 2) and forks positioning, we can assume that our formalism well captures replicon positioning and lengths observed in vivo.
As this study does not include any additional exploration or variation of the parameters inferred by Arbona et al. (ref. [63]), we consider a quantitative comparison with the nanopore replicon data to be beyond the scope of this paper.
Minor Comments:
The paper is in most places easy to follow. However, Section C bucked this trend and in general was quite difficult to follow. We would recommend that the authors try to revise this section to make clearer the actual physical parameters that govern a 'replication wave' and the formation of replication foci - how many forks, the extent to which the sisters are coordinated, etc for early vs. late replicating regions.
We now state more clearly with a sentence in the main text the driving forces behind the formation of such a “replication wave”. We believe that the several additions and clarifications following the various comments, improved the clarity of the manuscri
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In their manuscript, "Genome-wide modeling of DNA replication in space and time confirms the emergence of replication specific patterns in vivo in eukaryotes," authors Asaro et al perform computational modeling analyses to address an important open question in the chromatin field: how is DNA replication timing coupled to 3D genome architecture? Over the past ten years, the convergence of high-resolution replication timing (RT) analysis with high-resolution 3D genome mapping (e.g. 'Hi-C' technology) has resulted in the discovery that replication timing domains overlap considerably with 3D genomic domains such as topologically associating domains (TADs). How and why this happens both remain unknown, and advances in 3D genome mapping technology have provided even more data to model the problem of both 1) scheduling replication from distinct series of origins / initiation zones, and 2) modeling how 3D genome architecture is altered by the progression of replication forks, which inherently destroy chromatin structure before faithfully reforming G1 structures on daughter chromatids. As such, the problem being tackled by this computational manuscript is interesting.
Reviewer Comments / Significance
In their manuscript, "Genome-wide modeling of DNA replication in space and time confirms the emergence of replication specific patterns in vivo in eukaryotes," authors Asaro et al perform computational modeling analyses to address an important open question in the chromatin field: how is DNA replication timing coupled to 3D genome architecture? Over the past ten years, the convergence of high-resolution replication timing (RT) analysis with high-resolution 3D genome mapping (e.g. 'Hi-C' technology) has resulted in the discovery that replication timing domains overlap considerably with 3D genomic domains such as topologically associating domains (TADs). How and why this happens both remain unknown, and advances in 3D genome mapping technology have provided even more data to model the problem of both 1) scheduling replication from distinct series of origins / initiation zones, and 2) modeling how 3D genome architecture is altered by the progression of replication forks, which inherently destroy chromatin structure before faithfully reforming G1 structures on daughter chromatids. As such, the problem being tackled by this computational manuscript is interesting.
Asaro et al focus on the problem of how genome structure is altered by the progression of replisomes through S-phase in the budding yeast S. cerevisiae. The authors employ computational polymer modeling of G1 chromosomes, then implement a hierarchical model of replication origin firing along these polymers to examine how the G1 chromosome structural state is perturbed by replisome progression. Their results indicate that replication origins create 'fountains' - Hi-C map features that other groups have demonstrated are likely to originate from symmetric extrusion by condesin / cohesin complexes originating at a fixed point. These 'fountains' appear to be cohesin-independent, as revealed by depletion Hi-C experiments. Finally, the authors provide evidence from their model of a 'replication wave' that emanates from the spindle pole body. This is an interesting manuscript that raises some exciting questions for the field to follow up on.
Major Comments
Minor Comments:
Asaro et al focus on the problem of how genome structure is altered by the progression of replisomes through S-phase in the budding yeast S. cerevisiae. The authors employ computational polymer modeling of G1 chromosomes, then implement a hierarchical model of replication origin firing along these polymers to examine how the G1 chromosome structural state is perturbed by replisome progression. Their results indicate that replication origins create 'fountains' - Hi-C map features that other groups have demonstrated are likely to originate from symmetric extrusion by condesin / cohesin complexes originating at a fixed point. These 'fountains' appear to be cohesin-independent, as revealed by depletion Hi-C experiments. Finally, the authors provide evidence from their model of a 'replication wave' that emanates from the spindle pole body. This is an interesting manuscript that raises some exciting questions for the field to follow up on.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The paper "Genome-wide modelling of DNA replication in space and time confirms the emergence of replication specific patterns in vivo in eukaryotes" by D'Asaro et. al presents new computational and experimental results on the dynamics of genome replication in yeast. The authors present whole-nucleus scale simulations using a kinetic Monte Carlo polymer physics model. New HiC data for synchronised yeast samples with different protein knock-downs are also presented.
The main questions which the paper addresses are whether sister forks remain associated during replication, whether there is more general clustering of replication forks, and whether replication occurs in a 'spatial wave' through the nucleus. While the authors' model data are not able to conclusively show whether sister forks remain co-localised, the work provides some important insights which will be of high interest to the field.
I have no major issues with the paper, only some minor comments and suggestions to improve the readability of the manuscript or provide additional detail which will be of interest to readers. I list these here in the order in which they appear in the paper. There are also a number of typos and grammatical issues through the text, so I recommend thorough proofreading.
The work has been conducted thoroughly, and in general the paper is well written with good attention to detail. As far as I am aware, this is the first study where replication is simulated in a whole nucleus context, and the scale of the simulations is impressive. This allows the authors to address questions on replication foci and the spatiotemporal organisation of replication which would not be possible with more limited simulations, and to compare the model with previous experimental work. This, together with the new HiC data, I think this makes this a strong paper which will be of interested to biophysics and molecular biology researchers; the manuscript is written such that it would suit a interdisciplinary basic research audience.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript by D'Asaro et al. investigates the relationship between DNA replication and chromatin organization using polymer simulations. While this is primarily a simulation-based study, the authors also present relevant comparisons with experimental data and explore mechanistic aspects of replication fork interactions.
The primary weakness is that many aspects are not clear from the manuscript. Below is a list of questions that the authors must clarify:
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
By developing a new genome-wide 3D polymer simulation framework, D'Asaro et al. investigated the spatiotemporal interplay between DNA replication and chromatin organization in budding yeast: (1) T The simulations recapitulate fountain-like chromatin patterns around early replication origins, driven by colocalized sister replication forks. These findings align with Repli-HiC observations in human and mouse cells, yet the authors advance the field by demonstrating that these patterns are independent of Cohesin and Ctf4, underscoring replication itself as the primary driver. (2) Simulations reveal a replication "wave" where forks initially cluster near the spindle pole body (SPB) and redistribute during S-phase. While this spatial reorganization mirrors microscopy-derived replication foci (RFis), discrepancies in cluster sizes compared to super-resolution data suggest unresolved mechanistic nuances. (3) Replication transiently reduces chromatin mobility, attributed to sister chromatid intertwining rather than active forks. This work bridges replication timing, 3D genome architecture, and chromatin dynamics, offering a quantitative framework to dissect replication-driven structural changes. This work provides additional insights into how replication shapes nuclear organization and vice versa, with implications for genome stability and regulation. However, the following revisions could strengthen the manuscript:
Major:
In Sccl- and Ctf4-depleted cells, fountain signals persist or even intensify (Fig. 3A). This counterintuitive result warrants deeper investigation. Could the authors provide any suggestions or discussions? Potential explanations may include: Compensatory mechanisms (e.g., other replisome proteins stabilizing sister-forks). Altered chromatin mobility in mutants, enhancing Hi-C signal resolution. Artifacts from incomplete depletion (western blots for Sccl/Ctf4 levels should be included). 5. Inconsistent Figure References Several figure citations are mismatched. For instance, Fig. S1A has not been cited in the manuscript. Moreover, there is no Fig.1E in figure 1, while it has been cited in the text. All figure/panel references must be cross-checked and corrected.
Minor:
General assessment:
This study provides a valuable computational framework for investigating the dynamic interplay between DNA replication and 3D genome architecture. While the current implementation focuses on Saccharomyces cerevisiae, whose genome organization differs significantly from mammalian systems.
Advance: providing the first in vivo experimental evidence in investigating the role(s) of Cohesin and Ctf4 in the coupling of sister replication forks.
Audience: broad interests; including DNA replication, 3D genome structure, and basic research
Expertise: DNA replication and DNA damage repair within the chromatin environment.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
'The authors do not wish to provide a response at this time.'
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript by Quétin et al "Transient hypoxia followed by progressive reoxygenation is required for efficient skeletal muscle repair through Rev-ERBα modulation" describes the nature of muscle stem cell (MuSC) differentiation within its hypoxic niche using in vivo, ex vivo and in vitro methodologies. Approaches to limit oxygen in a regenerating model of muscle injury showed that muscle oxygenation is necessary for proper muscle repair. They found that the lack of oxygen is associated with the formation of hypotrophic myofibers, due to the inability of MuSCs to differentiate and fuse. Their findings show that the phenotype was independent of HIF-1α. However, RNA-seq of MuSCs 7 day post injury from prolonged hypoxia was shown to have significantly increased circadian clock gene Rev-erbα expression. Pharmacological inhibition of Rev-erbα during hypoxia rescued the myogenic phenotype. Contrarily, the use of Rev-erbα agonist in normoxia impaired the fusion capacity of MuSCs and decreases the number of large mature myofibres. This manuscript is well written and very easy to follow. Though, there are certain shortcomings outlined below. Sometimes the evidence provided does not support the conclusions made. For example, more rigour should be performed to state that there is a self-renewal phenotype.
Major issues
b) Moreover, the endothelial cell staining (Fig. 1B) appears to be unchanged in the time course of injury. To prove vascular damage this data should be corroborated, for example with lectin perfusion. 6. Problems with Figure 3J. There are data points with zero clusters/isolated myofibres suggesting that the hypoxic environment caused MuSCs to not activate from quiescence. There are several outliers for example at 1% there is a zero reading that makes the data significant. 7. In Figure 1G, Loxl2 after 14 days appears to be significant, as the error bars at 0 and 14 days do not overlap and thus it does not return to normal. An n=3 is not sufficient, as one of the data points at 14 days appears to be an outlier (the data stretching from 1500 to 3000). 8. In Fig. 2C and 2D, there are no control CSA and myofiber diameter experiments for keeping the mice in hypoxia over 14 and 28 days without injury. 9. For Figure 3K, how can self-renewing MuSCs be distinguished from MuSCs that never activated? Especially in the 1% O2 condition where few clusters formed. How does hypoxia influence activation? A 4hr or 8hr timepoint is necessary, as well as 24hrs. Also, for Figure 5E and 5F, it is possible that HIFcKO allowed the cells to activate normally, thus explaining the shift from quiescence to activation in the read-outs. This further highlights the importance of analyzing earlier timepoints. One cannot state that these cells are self-renewing or returning to quiescence without performing experiments on earlier timepoints. 10. The data for Figure 4 does not suggest that transient reoxygenation is required "for proper skeletal muscle repair" as stated by the authors only that reoxygenation has rescued the phenotype in the primary myoblasts. There is no hypoxia in the control (8% O2) for regeneration to occur (Fig. 2B). 11. One cannot rule out metabolic dysregulation. It's true that glycolytic fibers are generally larger than oxidative, it is likely that that alone does not explain the difference in fiber size. However, the fact that the fibers are more glycolytic does suggest a metabolic shift in the muscle (which was the aim of the experiment), which could also shift MuSC character altering their behaviour. How are MuSCs metabolically responding to hypoxia? 12. In Figure 2, how can one be sure that reoxygenation is blocked by the hypoxic chamber? Reduced O2 levels will induce hypoxia, but one cannot state that it blocks reoxygenation without further validation such as using pimonidazole as in Fig. 1E. If reoxygenation is blocked, then pimonidazole staining should remain consistent throughout the injury. 13. For Figure 3G, is a sum appropriate for the graph? Proportions would be more appropriate as cell number is not equal as shown in figure 3E. Can Pax7+/MyoD+ be defined as differentiated? By day 7, many MuSCs will have fused and be expressing MyoG, which is not accounted for by these definitions. Did systemic hypoxia increase self-renewal or impair activation? How can you distinguish these two? 14. In Figure 6A, while it is interesting that Pax7 levels are elevated in hypoxia and differentiation and fusion markers are down at 7days, it does not necessarily mean that self-renewal is increased. It might suggest that the hypoxic cells might have never activated or might have differentiated precociously. Are any cell cycle genes down regulated? Any other genes involved in quiescence altered? 15. The use of pimonidazole in Fig. 1E shows the staining within fibers (many with centrally located nuclei). These nuclei are differentiating and not representative of expanding MuSCs. How do the authors reconcile these MuSCs as part of their population.
Minor Problems
Referees cross-commenting
I agree with the thoughtful reviews and issues raised by Reviewers 1 and 2. I do not have anything more to add.
General Assessment: This manuscript is well written and easy to follow. It rigorously investigates the influence of oxygenation on MuSC behaviour. The authors utilize in vivo, ex vivo, and in vitro models to support their study and executed their work to a high degree. A limitation is that all experiments are only performed in mice and might not be applicable in humans. In addition, some claims made by the authors were over-reaching. The study can be improved by further validating some of the authors' claims, as has been suggested in the review.
Advance: This study is the first to report the effect of hypoxia on MuSCs in an ex vivo culture and in vivo injury model using a hypoxia chamber. This study helps clarify the role of HIF-1α on MuSC behaviour by suggesting that it does have a role in MuSC fate decisions. Finally, the authors make a novel link between circadian rhythm and MuSC behaviour in hypoxia.
Audience: A specialized audience that is interested in myogenesis, muscle stem cells, and/or hypoxia will be interested in this study. It highlights the important role of oxygen in muscle regeneration and may help researchers understand the role of oxygen in MuSC fate decisions.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript, Transient hypoxia followed by progressive reoxygenation is required for efficient skeletal muscle repair through Rev-ERBa modulation, revisits the role of hypoxia in skeletal muscle regeneration after acute injury. They first nicely demonstrate, using the pimonidazole hypoxia probe, that during regeneration skeletal muscle is transiently hypoxic at 5 days post injury (DPI). Then they show skeletal muscle regeneration is impaired in mice housed in a hypoxic (10% 02) chamber; the regenerated muscle mass is smaller, due to smaller regenerated myofibers and there is a shift in myofiber type so that there are more IIB myofibers. In addition, at 7 DPI when mice are raised in a hypoxic environment there is a shift in muscle stem cells so that they are more proliferative and fewer have differentiated. Ex vivo experiments culturing muscle stem cells in association with EDL myofibers in 1% 02, as compared with 8% 02, also led to fewer differentiated Pax7-MyoD+ cells, but could be restored if 02 was subsequently increased to 8%. They also found that low oxygen inhibited myoblast fusion in vitro. They then tested, via Pax7CreERT2/+;HIF-1afl/fl, whether HIF-1a signaling mediated the response of muscle stem cells to hypoxia in vivo. Surprisingly, they found that loss of HIF-1 did not impair myofiber regeneration in normoxic or hypoxic conditions, but they do provide some data suggesting that HIF-1a is required for the hypoxic-induced increase in Pax7+MyoD- muscle stem cells. Bulk RNA-seq analysis of 7 DPI muscle from mice housed in normoxic versus hypoxic conditions uncovered the interesting mis-regulation of circadian rhythm associated genes - in particular, the circadian clock repressor Rev-ERBa. Using a pharmacological antagonist of Rev-ERBa they show in culture that blocking Rev-ERBa (in contrast to loss of HIF-1a) rescues the fusion defect of muscle stem cells cultured in 1% 02. Conversely, they show that a Rev-ERBa agonist inhibits fusion in 8% 02. Altogether, the paper provides interesting new data on the controversial role of hypoxia and HIF-1a as well as data suggesting a connection between hypoxia and circadian rhythm genes. The data is logical and well presented, and the paper will be of strong interest to the regeneration and skeletal muscle research communities. I have two major comments and a list of smaller suggestions to improve the manuscript.
Major comments:
In vivo experiments (presented in Figures 2, 3, 5, 6, 7) house mice in hypoxic (10% oxygen) chambers, and the authors suggest that this blocks the progressive reoxygenation of skeletal muscle during regeneration. Surprisingly, the authors do not test when the mice are in hypoxic chambers whether, in fact, skeletal muscle is hypoxic at homeostasis and whether during regeneration muscle experiences prolonged hypoxia. The obvious experiment would be to use the pimonidazole probe on skeletal muscle sections of muscle at homeostasis and at 0, 5, 6, 14, and 28 DPI CTX injury in mice housed in hypoxic chambers. Without some demonstration that skeletal muscle oxygenation is changed when the mice are housed in hypoxic chambers, it is impossible to interpret these experiments.
The authors claim that reducing reoxygenation by maintaining the mice under systemic hypoxia impairs skeletal muscle repair by limiting the differentiation and fusion capacity of MuSCs in HIF-1a-independent manner, while it favors their return into quiescence through HIF-1a activation. They provide some in vitro evidence that Hif1ais required for the high levels Pax7+MyoD- muscle stem cells in 1% O2. They should also show that the elevated levels of Pax7+ muscle stem cells at 7 DPI (seen in Fig. 3D-G) requires HIF1a via analysis of Pax7CreERT2/+;HIF-1afl/fl mice.
Minor comments:
Please provide a reference for the pimonidazole probe. Reference 26, Hardy et al., is not the right one.
Please provide references that Loxl-2, Pdgfb, and Ang2 are HIF-inducible target genes.
Fig. 2C shows changes in average myofiber diameter. How was this calculated? Is this the largest diameter? Is there a reason that cross-sectional area was not measured (the more standard measurement)? Also, generally this type of data is shown as bar graphs - which is how these data are shown in Fig. 5C. Please also show the data in Fig. 2C as bar graphs.
Please provide reference for 8% 02 being physioxia in culture.
Fig.5 should also quantify the number of centronuclei/myofiber (as in Fig. 2I) for Pax7CreERT2/+;HIF-1afl/fl mice 14 and 28 DPI - to further demonstrate that differentiation defects in hypoxia are HIF-1a independent.
Please provide a graphical model of your research findings.
There are many typos and verb tense issues. Please fix these. The most amusing is Stinkingly in the Discussion.
Referees cross-commenting
I think several important issues are raised by myself and reviewer 3. First, the authors need to explain and support their use of 10% O2 hypoxia in vivo chambers and 1% O2 for hypoxic in vitro experiments. Second, the authors have not demonstrated that reoxygenation of muscle is prevented in mice raised in hypoxic chamber. There are questions about how well the pimonidazole probe is working (the widespread expression at 5 dpi in Fig. 1E suggests there may be specificity issues) and this probe is also not shown for muscle from mice living in hypoxic chambers. Another method of demonstrating hypoxia in muscle tissue would be useful.
The paper provides interesting new data on the controversial role of hypoxia and HIF-1a as well as data suggesting a connection between hypoxia and circadian rhythm genes.
This paper will be of interest to researchers studying the role of hypoxia on regeneration and also to researchers studying muscle regeneration.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
SUMMARY
Quétin et al investigated the dynamics of oxygen levels during the skeletal muscle regeneration following sterile damage and its impact on muscle repair. They combined in vivo and ex-vivo model systems, together with genetic and pharmacological manipulations. They found results consistent with the fact that a dynamic oxygeneation process, hypoxia during the early phase followed by reoxygenation, are involved in muscle repair. Prolonged hypoxia leads to defective myogenesis and muscle repair. These activities apper to be meadiated by modulation of Rev-ERBα levels. Collectively, the study provide intriguing insight regarding the role of oxygen in muscle repair.
MAJOR COMMENTS
In Figure 1, the 5 days post CTX injury is too late to claim that "myogenic cell expansion is initiated in a hypoxic environment". Indeed, at day 5 myofibers are already regenerated, although immature. To support their claim, the authors should perform analyses and quantification of Pax7+, Pax7+Ki67+ and hypoxia at earlier timepoints.
In Figure 2B, a larger number of mononuclear cells is present in hypoxia mice. Is hypoxia affecting the number/activity of extra-muscular cells important for muscle regeneration like for example FAPs, macrophages, etc?
In Figure 5H, the myotubes formed by HIF-1α cKO appear thinner than control myotubes. Is myotube size affected by lack of HIF1 α?
The choice of the 7 days post CTX for the RNA-seq is odd. Indeed, at that timepoint there are obvious histological abnormalities in hypoxia mice. Hence, it is highly likely that many DEGs are simply secondary to the defect in regeneration and not directly linked to hypoxia exposure. This is probably the reason why the authors found so many (close to 4K) DEGs. To focus on the genes closely-associated to the primary defect, the authors should have performed the RNA-seq at an earlier timepoint, in which minimal histological defects were present. While repeating the RNA-seq would be costly and time consuming, the authors could at least address this issue by RT-qPCR. Are muscle stem cell fate, repair, and circadian clock genes significantly altered 3 and 5 days after CTX injury in hypoxia vs normoxia?
Given that compounds have frequently off-target effects, the authors must independently support their Rev-ERBα findings by performing genetic manipulations, at least ex-vivo.
A recent study (PMID: 38333911), which was not cited by the authors, reports muscle atrophy and weakness, impaired muscle regeneration, and increased fibrosis in hypoxia exposed mice. Intriguingly, this was due to impaired MuSC proliferation and differentiation following HIF-2α stabilization under hypoxia. Hence, the authors should investigate if HIF-2α plays any role in the phenotypes they describe. For example, is HIF-2α a regulator of circadian clock genes expression?
Referees cross-commenting
The other reviewers raised very relevant issues and I fully agree with their comments. In particular, I concur with Reviewer #3 that in several instances the evidence provided by the authors does not support the conclusions made.
SIGNIFICANCE
There is a limited knowledge regarding the role of oxygen supply during tissue differentiation and repair. In the muscle field, there are conflicting reports in the literature. This study combines genetic, pharmacological and oxygen manipulations both in vivo and ex-vivo to investigate the role of oxygen during regeneration following sterile skeletal muscle injury. The results are very intriguing and potentially relevant both for muscle, but possibly also for other tissue repair. Aspects of the study that must be improved concern the role of HIF-1a and HIF-2α in the process, and the characterization of the molecular mechanism through which Rev-ERBα is regulated by oxygen and regulates muscle repair.
AUDIENCE: specialized, basic research, translational research; results could potentially extend beyond the muscle field.
FIELD OF EXPERTISE: muscle differentiation, muscular dystrophy, gene expression regulation.
Download the complete Review Process [PDF] including:
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
The authors do not wish to provide a response at this time.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
SUMMARY OF THE PRESENTED FINDINGS
Abstract
Introduction
a. These structures facilitate endosomal escape due to protonation of tertiary amines at lower pH7.
Results
b. In human models MDAMB-231 and MCF7 cells, the NPs also showed high eGFP mRNA transfection efficiency
c. The efficiency of eGFP mRNA-loaded pBAE-NPs to transfect mRNA into different mouse breast cancer cells (AT3, 4T07, EO771, EMT6, 66cl4, EpRAS, and 4T1) was tested using NPs encapsulating eGFP mRNA,
d. Synthetic Lcor mRNA contained a Cap1, 5' and 3' untranslated regions (UTR) and a standard polyA tail (Fig. S2A), and all uracil were replaced for 5-methoxyuracil (5-moU) to avoid immunogenic reactions27,28. First, we measured and detected high levels of Lcor mRNA by qRT-PCR
e. NPs were stable at 25ºC for 24 h (Fig. S2C). In contrast, under conditions simulating the physiological environment (37ºC), a decrease in FRET signaling was detected ... indicating disassembly of the NPs after 2 h (Fig. S2C).
f. Lcor mRNA NPs, induces the expression of APM genes in AT3 and 4T07 cell lines
g. AT3 cells that constitutively overexpress ovalbumin (OVA). In these cells, OVA is cleaved, generating the SIINFEKL antigen peptide presented in the H-2Kb context. This can be used to measure APM activity using the anti-SIINFEKL antibody via flow cytometry.
h. We also observed a time- and dose-dependent effect regarding APM induction.
i. When tumors reached 0.5 x 0.5 cm2, we treated them intratumorally with pBAE-NPs loaded with 5 ug of synthetic FLuc or eGFP mRNA. We detected BLI at 3 h, meaning that tumor cells had taken up the mRNA-loaded NPs and translated a luciferase active protein within 3 h. In both models, expression peaked around 6 to 10 hours after administration
j. After local administration of 5 μg of Lcor mRNA-loaded NPs, we observed a rapid increase in Lcor mRNA in the tumor tissue, followed by a decrease, reaching baseline levels after 24 h (Fig. 3C). ..To unravel the protein dynamics, we used ... LCOR-HA protein and uniquely detect the ectopic protein using anti-HA by IF. As expected, LCOR-HA protein expression was delayed, peaking 3 h after administration (Fig. 3D). Linked to protein expression, at 3 h and 6 h after administration, we detected an increase in APM genes by RT-qPCR (Fig. 3E and S3D).
k. the combination of Lcor mRNA-loaded NPs with anti-PDL1 therapy not only reduced tumor growth but also led to tumor eradication in 5 out of 7 mice.
l. The combination of Lcor mRNA-loaded NPs with different ICIs showed high efficiency in preclinical models, thus supporting the feasibility of starting clinical studies and thus bringing the treatment closer to patients.
Major points
L. 277: "NPs were stable at 25ºC for 24 h (Fig. S2C). In contrast, under conditions simulating the physiological environment (37ºC), a decrease in FRET signaling was detected ... indicating disassembly of the NPs after 2 h (Fig. S2C)." - The disassembly of the NPs after 2 h is key to the performance of the chosen approach.
L. 296: "The results showed an increased number of cells with higher OVA-SIINFEKL presentation, indicating the enhanced activity of the APM induced by the Lcor mRNA-loaded pBAE-NPs... demonstrate the efficiency of this mRNA nanotechnology to rescue the function of the LCOR TF in inducing tumor cell immunogenicity and thus modulating tumor phenotypes." - There is a key difference between activating antigen-presenting machinary and inducing immunogenicity, i.e. recognition by the immune system and activation of effector cells. There is no indication on how effective endogenous immune responses (e.g. antibody titers, TIL infiltration, cytokine release) are to the administration of Lcor mRNA-loaded NPs.
L. 325: "Based on these results, we estimated an optimal therapeutic regimen of Lcor-mRNA-loaded pBAE-NPs administration in our preclinical experimental models would be every 3 days." - It is highly unclear how the authors came to this conclusion, as it should be based on the time frame of optimal immune responses.
L. 332: "Lcor mRNA-loaded NPs were administered at a dose of 250 μg/kg by intratumoral (i.t.) injection twice a week" - This possibly is the strongest limitation of this study. Intratumor injections of largely unfeasible/unrealistic in clinical setting. Even more, the management of metastatic disease appears out of question.
L. 337: "the results revealed that Lcor mRNA monotherapy was enough to reduce 4T07 tumor 338 growth." - These effects appear rather limited (Fig. 4A,B) and are not statistically significant in Fig. S4B and Fig. S5A.
L. 338: "the combination of Lcor mRNA-loaded NPs with anti-PDL1 therapy not only reduced tumor growth but also led to tumor eradication in 5 out of 7 mice" - Fig. 4A bottom left panel. Three of the tumor growth curves abruptly stop at below 200 mm3. Typically, this is mouse death. This reduces the tumor pool to four xenografts. Among these, we notice two complete responses and two tumor progressions. Two tumor progressions are seen also in the combination Lcor mRNA+ α-PD-L1 group. We are unsure about the statistics of this experiment.
L. 350: "The combination of Lcor mRNA-loaded NPs with different ICIs showed high efficiency in preclinical models, thus supporting the feasibility of starting clinical studies and thus bringing the treatment closer to patients."
General assessment:strengths and limitations.
The identification of a candidate therapeutic means, by supplying Lcor mRNA for induction of antigen-presenting molecules is of potential interest. As this is not a basic science study, but aims at developing feasible therapeutics, it falls short in this respect, as most likely unfeasible in patients. The combined effect with anti-immune blockade agents is of interest. However, if one assumes that effective immunostimulation was indeed induced by Lcor mRNA, its overall impact on tumor growth is per se weak, if any. Maybe only antigen presentation is induced, but this is in the absence of costimulatory signals? This needs to be investigated.
Advance
This article is based on good papers that were published years ago. The science novelty is limited. As the idea is to develop a novel therapeutic approach, the lack of realistic feasibility severely limits merits.
Audience
Scientists involved in preclinical studies.
Reviewer expertise
This reviewer and his research group have cloned the genes and biochemically characterized novel tumor drivers. He identified their function as stimulators of tumor cell growth and of metastatic spreading, together with roles in cell-cell adhesion, signal transduction and local cancer invasion. This led to the discovery of their prognostic / predictive relevance in human cancer. Two murine models of rare genetic diseases were generated by ablating the corresponding murine genes. He then pioneered the development of software for the identification of fusion oncogenes and of transcription factor-DNA binding sites. This reviewer fostered novel anti-cancer immunotherapies. He generated anti-cancer cytotoxic T lymphocytes, by the use of in vitro engineered antigen presenting cells. Using proprietary discovery platforms, this reviewer developed novel anti-cancer monoclonal antibodies, that selectively target cancer cells. This led to the engineering of humanized antibody-drug conjugates, bispecific anti-CD3/activated Trop-2 antibodies and innovative CAR-T designs. ADCs are now being tested in clinical trials in cancer patients.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this manuscript, Serra-Mir et al investigate the therapeutic potential of delivering the mRNA of LCOR transcription factor via nanoparticles to enhance the efficacy of immune checkpoint inhibitors. The authors show that the mRNA delivery mediated by H and R-nanoparticles was efficient in multiple breast cancer cell lines in vitro. Moreover, using mouse models, they show that LCOR mRNA delivery may improve the efficacy of the treatment with anti-PDL1 or anti-CTLA4 checkpoint inhibitors against tumors. Although this proof-of-concept study has promising aspects, there are significant weaknesses that should be addressed. Details below.
Major points:
Minor points:
The study is a proof-of-concept investigation addressing whether LCOR mRNA can be delivered by nanoparticles to sensitize tumors to immunotherapy. This approach aims to overcome the limitations and difficulties of targeting transcription factors for therapeutic purposes. However, although the delivery of LCOR mRNA appears to be sufficient, further characterization of the resulting impact needs to be done. This includes both impact on immune responses as well as cell-autonomous impact on cancer cell proliferation and apoptosis.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Manuscript number: RC-2025-02946
Corresponding author(s): Margaret, Frame
Roza, Masalmeh
[Please use this template only if the submitted manuscript should be considered by the affiliate journal as a full revision in response to the points raised by the reviewers.
If you wish to submit a preliminary revision with a revision plan, please use our "Revision Plan" template. It is important to use the appropriate template to clearly inform the editors of your intentions.]
This section is optional. Insert here any general statements you wish to make about the goal of the study or about the reviews.
We thank the reviewers for recognizing the significance of our work and for their constructive feedback and suggestions, most of which we have implemented in our revised manuscript.
This section is mandatory. *Please insert a point-by-point reply describing the revisions that were already carried out and included in the transferred manuscript. *
Reviewer #1
Evidence, reproducibility and clarity
Review of Masalmeh et al. Title: "FAK modulates glioblastoma stem cell energetics..."
Previous studies have implicated FAK and the related tyrosine kinase PYK2 in glioblastoma growth, cell migration, and invasion. Herein, using a murine stem cell model of glioblastoma, the authors used CRISPR to inactivate FAK, FAK-null cells selected and cloned, and lentiviral re-expression of murine FAK in the FAK-null cells (termed FAK Rx) was accomplished. FAK-/- cells were shown to possess epithelial characteristics whereas FAK Rx cells expressed mesenchymal markers and increased cell migration/invasion in vitro. Comparisons between FAK-/- and FAK Rx cells showed that FAK re-expressed increased mitochondrial respiration and amino acid uptake. This was associated with FAK Rx cells exhibiting filamentous mitochondrial morphology (potentially an OXPHOS phenotype) and decreased levels of MTFR1L S235 phosphorylation (implicated in mito morphology fragmentation). Mito and epithelial cell morphology of FAK-/- cells was reversed by treatment with Rho-kinase inhibitors that also increased mito metabolism and cell viability. Last, FAK-dependent glioblastoma tumor growth was shown by comparisons of FAK-/- and FAK Rx implantation studies.
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
Some questions that would enhance potential impact. 1. Cell generation. Please describe the analysis of FAK-/- clones in more detail. The "low viability" phenotype needs further explanation with regard to clonal expansion and growth characteristics?
Response:
Figure 1F: need further support of MET change upon FAK KO and EMT reversion.
Response: We have added a heatmap (Figure S1E) illustrating the changes in protein expression of core-enriched EMT/MET genes products (by proteomics) after FAK gene deletion (EMT genes as defined in Howe et al., 2018) ; this strengthens the conclusion that the MET reversion morphological phenotype is accompanied by recognised MET protein changes.
Fig. 2: Need further support if FAK effects impact glycolysis or oxidative phosphorylation in particular as implicated by the stem cell model.
Response: We show that FAK impacts both glycolysis (Figure 2A, 2E, and 2F) and mitochondrial oxidative phosphorylation on the basis of the oxygen consumption rate (OCR) (Figure 2B, and 2D), showing both are contributing pathways to FAK-dependent energy production. We have clarified this in the text.
Is there a combinatorial potential between FAKi and chemotherapies used for glioblastoma. Need to build upon past studies.
Response: Yes, previous studies suggest that inhibiting FAK can sensitize GBM cells to chemotherapy (Golubovskaya et al., 2012; Ortiz-Rivera et al., 2023). We have included a paragraph in the discussion section to make sure this is clearer. Although it is not the subject of this study, we appreciate it is useful context.
The notation of changes in glucose transporter expression should be followed up with regard to the potential that FAK-expressing cells may have different uptake of carbon sources and other amino acids. Altered uptake could be one potential explanation for increase glycolysis and glutamine flux.
Response: We agree with the reviewer that glucose uptake could be contributing and we include data that 2 glucose transporters are indeed FAK-regulated namely Glucose transporter 1 (GLUT1, encoded by Slc2a1 gene) and Glucose transporter 3 (GLUT 3, encoded by Slc2a3 gene) (shown in Figure S2B and C).
It would be helpful to support the confocal microscopy of mitos with EM.
Response:
We are concerned (and in our experience) that Electron microscopy (EM) may introduce artefacts during sample preparation. In contrast, immunofluorescence sample preparation is less susceptible to artefacts. The SORA system we used is not a conventional point-scanning confocal microscope, but is a super-resolution module based on a spinning disk confocal platform (CSU-W1; Yokogawa) using optical pixel reassignment with confocal detection. This method enhances resolution in all dimensions with resolution in our samples measured at 120nm. This has been instructive in defining a new level of changes in mitochondrial morphology upon FAK gene deletion.
Lack of FAK expression with increased MTFR1 phosphorylation is difficult to interpret.
Response: We do not directly show that this phosphorylation event is causal in our experiments; however, we think it important to document this change since it has been published that phosphorylation of MTFR1 has been causally linked to the mitochondrial morphology we observed in other systems (Tilokani et al., 2022).
Need to have better support between loss of FAK and the increase in Rho signaling. Use of Rho kinase inhibitors is very limited and the context to FAK (and or Pyk2) remains unclear. Past studies have linked integrin adhesion to ECM as a linkage between FAK activation and the transient inhibition of RhoA GTP binding. Is integrin signaling and FAK involved in the cell and metabolism phenotypes in this new model?
Response: To better support the antagonistic effect of FAK on Rho-kinase (ROCK) signalling, we included a new experiment in which the integrin-FAK signalling pathway has been disrupted by treating FAK WT cells with an agent that causes detachment from the substratum, Accutase, and growing the cells in suspension in laminin-free medium. We present ROCK activity data, as judged by phosphorylated MLC2 at serine 19 (pMLC2 S19), relating this to induced FAK phosphorylation at Y397 (a surrogate for FAK activity) that is supressed after integrin disengagement. These measurements have been compared with conditions whereby integrin-FAK signalling is activated by growing the cells on laminin coated surfaces. We observed a time-dependent decrease in pFAK(Y397) levels (normalised to total FAK) in suspended cells compared to those spread on laminin, while pMLC2(S19) levels increased in a reciprocal manner over time in detached cells relative to spread cells (S4A and B). There is therefore an inverse relationship between integrin-FAK signalling and ROCK-MLC2 activity, consistent with findings from FAK gene deletion experiments. In the former case, we do not rely on gene deletion cell clones.
Significance
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
__Response: __
Deleting the gene encoding FAK in mouse embryonic fibroblasts leads to elevated Pyk2 expression (Sieg, 2000). However, in the GBM stem cell model we used here, Pyk2 was not expressed (determined by both transcriptomics and proteomics). We have included Figure S1E to show that PYK2 expression was undetectable in FAK -/- and FAK Rx cells at the RNA level (Figure S1F). We conclude that there is no compensatory increase in Pyk2 upon FAK loss in these cells. In the transformed neural stem cell model of GBM, we do not consistently or robustly detect nuclear FAK.
Review #2
Masalmeh and colleagues employ a neural stem/progenitor cell-based glioma model (NPE cells) to investigate the role of Focal Adhesion Kinase (FAK) in GBM, with a focus on potential links between the regulation of morphological/adhesive and metabolic GBM cell properties. For this, the authors employ wt cells alongside newly generated FAK-KO and -reexpressing cells, as well as pharmacological interventions to probe the relevance of specific signaling pathways. The authors´ main claims are that FAK crucially modulates glioma cell morphology, cell-cell and cell-substrate interactions and motility, as well as their metabolism, and that these effects translate to changes to relevant in vivo properties such as invasion and tumor growth.
My main issues are with the model chosen by the authors.
As per the methods section, generation of FAK-KO and -"Rx" NPE cells entailed protracted selection/expansion processes, which may have resulted in inadvertent selection for cellular/molecular properties unrelated to the desired one (loss or gain of FAK expression) and which may have had cascading effects on NPE cells. The authors nonetheless repeatedly claim the parameters they quantify, such as mitochondrial or cytoskeletal properties or metabolic features, to have directly resulted from FAK loss or reintroduction. Examples of such causal inferences are to be found in lines 123, 134/135, 165, 181. Such causal claims are, in my view, unsupported.
Acute perturbation of FAK expression/activity, genetically or pharmacologically, followed by a rapid assessment of the processes under investigation, would be needed to begin to assess causality, even if acute genetic perturbations may be technically challenging as sufficient gene expression reduction or restoration to physiologically relevant levels may be hard to achieve.
Response:
We would like to first comment on the model we used here, which we think will clarify the validity of our approach. The model is a transformed stem cell model of GBM that was published in (Gangoso et al., Cell, 2021) and is now used regularly in the GBM field. As mentioned in the response to Reviewer 1, we have added text (page 4 and 5 in the revised manuscript) and a new supplementary figure (Figure S1D) clarifying that the morphological changes we observed were consistent across multiple FAK -/- clones, showing this was not due to any inter-clonal variability. We also added images showing that the morphological changes were apparent at 48 h after nucleofecting FAK -/- cells with the FAK‑expressing vector specifically (not the empty vector), prior to starting G418 selection to enrich for FAK‑expressing cells (Figure S1C), addressing the worry that clonal variation and selection was the cause of the FAK-dependent phenotypes we observed. We believe that our model provides a type of well controlled, clean genetic cancer cell system of a type that is commonly used in cancer cell biology, allowing us to attribute phenotypes to individual proteins.
We have also carried out a more acute treatment by using the FAK inhibitor VS4718 to perturb FAK kinase activity and assessed the effects on glycolysis and glutamine oxidation after 48h treatment (Figure S2D, E and F). We found that treating the transformed neural stem cells (parental population) with FAK inhibitor (300nM VS4718) decreases glucose incorporation into glycolysis intermediates and glutamine incorporation into TCA cycle intermediates, consistent with a role for FAK’s kinase activity in maintaining glycolysis and glutamine oxidation.
The employed pharmacological modulation of ROCK activity is the only approach that, given the presumably acute nature of the treatment, may have allowed the authors to probe the proposed functional links. The methods section of the manuscript does not however comprise details as to the duration of these treatments, which leaves open the possibility of long-term treatment having been carried out (data shown in Figure 5B refers to 72hr treatment).
__Response: __
We have added the duration of the treatment to the Methods section and Figure Legends, to clarify that cells were treated with ROCK inhibitors for 24h, before assessing the effects on mictochondria (Figure 4C, D, S4C and D) and glutamine oxidation (Figure 5A, and S5). For metabolic activity by AlamarBlue assay, cells were treated with ROCK inhibitors for 72h (Figure 5B).
Even in the case of ROCK inhibitor experiments, it is however unclear if and how the effects on cell morphology and adhesion, mitochondrial organization and metabolic activity may be connected to each other and, if at all, to FAK expression.
Given the above uncertainties due to the nature of the model and experimental approaches, it is hard to assess the reliability and thus the relevance of the findings.
Response:
FAK suppresses ROCK activity (as judged by pMLC2 S19, Figure 4A and B). Treating FAK -/- cells with two different ROCK inhibitors restored mesenchymal-like cell morphology, mitochondrial morphology and glutamine oxidation. As mentioned above, to strengthen our evidence for the antagonistic role of FAK in ROCK-MLC2 signalling, we have now introduced an experiment whereby integrin-FAK signalling was disrupted through treatment with a detachment agent (Accutase), and subsequently maintaining the cells in suspension in laminin-free medium. We assessed pMLC2 S19 levels (a measure of ROCK activity) relating this to FAK phosphorylation that is supressed after integrin disengagement. These results were evaluated relative to spread wild type cells growing on laminin where Integrin-FAK signalling was active (Figure S4A and B). We observed an inverse relationship between Integrin-FAK signalling and ROCK-MLC2 activity in keeping with our conclusions (Figure 4A and B).
Experimental support for the ability of cell-substrate interaction modulation to concomitantly impact cellular metabolism and motility/invasion would be significant both in terms of advancing our understanding of glioma cell biology and of its translational potential, but the evidence being provided is at best compatible with the proposed model.
Response: We carried out a new experiment to support the ability of cell-substrate interaction modulation to impact metabolism; specifically, we inhibited cell-substrate interactions by plating the cells on Poly-2-hydroxyethyl methacrylate (Poly 2-HEMA)-coated dishes. This suppressed FAK phosphorylation at Y397, as expected, with concomitant reduction in glutamine utilisation in the TCA cycle (Figure S3A, B and C).
My background/expertise is in developmental and adult neurogenesis, in vivo modelling of gliomagenesis and cell fate control/reprogramming, with a focus on molecular mechanisms of differentiation and quantitative aspects of lineage dynamics; molecular details of the control of cellular metabolism, cell-cell adhesion and cytoskeletal dynamics are not core expertise of mine.
We appreciate this reviewer’s expertise are not necessarily in the cancer cell biology and genetic intervention aspects of our study. We hope that the explanations we have provided satisfy the reviewer that our conclusions are valid.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Masalmeh and colleagues employ a neural stem/progenitor cell-based glioma model (NPE cells) to investigate the role of Focal Adhesion Kinase (FAK) in GBM, with a focus on potential links between the regulation of morphological/adhesive and metabolic GBM cell properties. For this, the authors employ wt cells alongside newly generated FAK-KO and -reexpressing cells, as well as pharmacological interventions to probe the relevance of specific signaling pathways. The authors´ main claims are that FAK crucially modulates glioma cell morphology, cell-cell and cell-substrate interactions and motility, as well as their metabolism, and that these effects translate to changes to relevant in vivo properties such as invasion and tumor growth. My main issues are with the model chosen by the authors.
As per the methods section, generation of FAK-KO and -"Rx" NPE cells entailed protracted selection/expansion processes, which may have resulted in inadvertent selection for cellular/molecular properties unrelated to the desired one (loss or gain of FAK expression) and which may have had cascading effects on NPE cells. The authors nonetheless repeatedly claim the parameters they quantify, such as mitochondrial or cytoskeletal properties or metabolic features, to have directly resulted from FAK loss or reintroduction. Examples of such causal inferences are to be found in lines 123, 134/135, 165, 181. Such causal claims are, in my view, unsupported. Acute perturbation of FAK expression/activity, genetically or pharmacologically, followed by a rapid assessment of the processes under investigation, would be needed to begin to assess causality, even if acute genetic perturbations may be technically challenging as sufficient gene expression reduction or restoration to physiologically relevant levels may be hard to achieve.
The employed pharmacological modulation of ROCK activity is the only approach that, given the presumably acute nature of the treatment, may have allowed the authors to probe the proposed functional links. The methods section of the manuscript does not however comprise details as to the duration of these treatments, which leaves open the possibility of long-term treatment having been carried out (data shown in Figure 5B refers to 72hr treatment). Even in the case of ROCK inhibitor experiments, it is however unclear if and how the effects on cell morphology and adhesion, mitochondrial organization and metabolic activity may be connected to each other and, if at all, to FAK expression.
Given the above uncertainties due to the nature of the model and experimental approaches, it is hard to assess the reliability and thus the relevance of the findings.
Experimental support for the ability of cell-substrate interaction modulation to concomitantly impact cellular metabolism and motility/invasion would be significant both in terms of advancing our understanding of glioma cell biology and of its translational potential, but the evidence being provided is at best compatible with the proposed model.
My background/expertise is in developmental and adult neurogenesis, in vivo modelling of gliomagenesis and cell fate control/reprogramming, with a focus on molecular mechanisms of differentiation and quantitative aspects of lineage dynamics; molecular details of the control of cellular metabolism, cell-cell adhesion and cytoskeletal dynamics are not core expertise of mine.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Review of Masalmeh et al.
Title: "FAK modulates glioblastoma stem cell energetics..."
Previous studies have implicated FAK and the related tyrosine kinase PYK2 in glioblastoma growth, cell migration, and invasion. Herein, using a murine stem cell model of glioblastoma, the authors used CRISPR to inactivate FAK, FAK-null cells selected and cloned, and lentiviral re-expression of murine FAK in the FAK-null cells (termed FAK Rx) was accomplished. FAK-/- cells were shown to possess epithelial characteristics whereas FAK Rx cells expressed mesenchymal markers and increased cell migration/invasion in vitro. Comparisons between FAK-/- and FAK Rx cells showed that FAK re-expressed increased mitochondrial respiration and amino acid uptake. This was associated with FAK Rx cells exhibiting filamentous mitochondrial morphology (potentially an OXPHOS phenotype) and decreased levels of MTFR1L S235 phosphorylation (implicated in mito morphology fragmentation). Mito and epithelial cell morphology of FAK-/- cells was reversed by treatment with Rho-kinase inhibitors that also increased mito metabolism and cell viability. Last, FAK-dependent glioblastoma tumor growth was shown by comparisons of FAK-/- and FAK Rx implantation studies.
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
Some questions that would enhance potential impact.
The studies by Masalmeh provide interesting findings associating FAK expression with changes in mitochondrial morphology, energy metabolism, and glutamate uptake. According to the authors model, FAK expression is supporting a glioblastoma stem cell like phenotype in vitro and tumor growth in vivo. What remains unclear is the mechanistic connection to cell changes and whether or not these are be dependent on intrinsic FAK activity or as the Frame group has previously published, potentially FAK nuclear localization. The associations with MTFR1L phosphorylation and effects by Rho kinase inhibition are likely indirect and remind this reviewer of long-ago studies with FAK-null fibroblasts that exhibit epithelial characteristics, still express PYK2, exhibited elevated RhoA GTPase activity. Some of these phenotypes were linked to changes in RhoGEF and RhoGAP signaling with FAK and/or Pyk2. At a minimum, it would be informative to know whether Pyk2 signaling is relevant for observed phenotypes and whether the authors can further support their associations with FAK-targeted or FAK-Pyk2-targeted inhibitors or PROTACs.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
'Please download the Response to Reviewers file, which contains Reviewers comments and authors responses and new data'
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this work, the authors investigate the cytoplasmatic roles of Mei2, an RNA-binding protein in fission yeast, in particular its interactions with processing bodies (PBs) in the cytoplasm. The manuscript rests heavily on microscopy data, using a combination of time-resolved microscopy and molecular mutation and tagging techniques.
Mei2 is known for its role in the nucleus of zygotic cells. Here, it is shown that Mei2 co-localizes with the PB markers Dcp2 and Edc3. This happens in zygotes but not in gametes (e.g. when fusion is blocked in fus1 mutants) (Fig 4E). <br /> This co-localization in PBs is counteracted by Pat1-driven phosphorylation of Mei2. Phosphorylation by Pat1 is known to suppress Mei2 activity. Mei3 inhibits Pat1; in a mei3 mutant Mei2 cannot accumulate in PBs, the same happens with a non-phosphorylatable mei2 allele (Fig. 5). In a pat1Δ mutant, constitutively active Mei2 is compatible with growth if it stays in the nucleus (mei2-NLS), but not if Mei2 is forced to the cytoplasm (mei2-NES) (Fig. 3G). This indicates that it is the cytoplasmic function of Mei2 that is critical.
Forcing Pat1 to be cytoplasmic (Pat1-NES) allowed normal vegetative growth and mating (Fig. 3A-C), whereas nuclear Pat1 (Pat1-NLS) produced premature mating (Fig. 3A,B). Thus, cytoplasmic Pat1 phosphorylation of Mei2 is critical for controlling the transition from mitotic growth to fusion and zygote formation.
Mei2 shuttles between the nucleus and cytoplasm, and one of its RNA-binding domains (RRM1) drives nuclear import, while both RRM1 and RRM3 are required for export to the cytoplasm (Fig. 2 and S2). Little was known previously of the role of RRM1.
They present evidence that this localization to PBs is required for development. Knocking out the RNA helicase Ste13 (ortholog of S. cerevisiae Dhh1 which is a PB component) reduces PB formation (Fig. 6A). Even a non-phosphorylatable mei2 allele (i.e. it cannot be inactivated by Pat1) is incapable of driving sporulation in a ste13Δ background (Fig. 6B-D). This demonstrates that Mei2 activity is dependent on PBs.
The study is well conceived and performed, and the conclusions mostly well backed by data. Experimental and statistical procedures are well described, and the number of replicates is sufficient.
There are some minor questions however:
In the literature, Mei2 is described as appearing as a nuclear dot in zygotic cells, but invisible in mitotic cells. Here, the authors demonstrate a Mei2 dot already 30 minutes before fertilization (Fig. 2A). Is the reason for this a more sensitive microscopic technique, or something else?
The authors claim that the RRM1 RNA-binding region of Mei2 is essential for cytoplasmic Mei2 function and recruitment to PBs. This contrasts with previous publications (Watanabe 1994, Watanabe 1997, Otsubo 2014), as pointed out by the authors, where RRM1 appears to be dispensable for development. How do the authors argue about this discrepancy?
Overall, this paper presents major advances in our understanding of the cytoplasmic functions of this intensely studied RNA-binding protein, Mei2, in the transitions between the mitotic and meiotic cell cycles.
It builds on the original observations of Mei2 as an essential protein for fusion and meiosis (Watanabe EMBO J 1988), being RNA-binding (Watanabe Cell 1994), and forming a nuclear dot in meiotic cells (Yamashita Cell 1998). These were followed by e.g. reports how Pat1 phosphorylation regulates Mei2 degradation (Matsuo J Cell Sci 2007) and its binding to RNA (Shen J Mol Cell Biol 2022). The present manuscript gives a broader view of the functions of Mei2 beyond its previously described role in the nucleus, and characterizes its interactions with the other players in fusion and meiosis.
These findings will be of great interest not only to the fission yeast community, but to a wide range of scientists specializing in meiosis and fertilization, and to the RNA biologists at large. Since Mei2 is conserved across many branches of the eukaryotic tree as an RNA-binding protein, albeit with somewhat different functions in e.g. plants, the work has general relevance.
I have read this manuscript with a background in general yeast cell and molecular biology, including post-transcriptional regulation. I am no microscopy expert, however I find the experimental setup with fluorescent tagging, combinations of mutations in key components in the pathway, and high resolution microscopy data from time series, convincing.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this manuscript, Araoyinbo et al. present a wealth of detailed data analyzing the cellular behavior of mainly three proteins, the RNA-binding protein Mei2, the kinase Pat1 and its inhibitor the protein Mei3 during mating and subsequent initiation of meiosis in fission yeast. This analysis involve also the detailed testing of potential models about how these protein act on each other to fulfil their different functions, such as to block remating on zygotes, the initiation of zygotic S-phase and the initiation of meiosis and sporulation. These data converge to a model whereby Pat1 inhibition by Mei3 expression upon cell fusion unleashes Mei2 function in the cytoplasm. This is due to the subsequent dephosphorylation of Mei2, and its RNA-recognition motif RRM1 interacting with and recruiting Mei2-bound RNAs to P-bodies, where their translation is most likely repressed (at least the translation of a synthetic mRNA - Mei2 pair is repressed when the pair is targeted to P-bodies). Together, this study provides detailed insights into how the meiotic cycle is induced upon mating of fission yeast cells but not in gametes).
Overall, this is a very carefully controlled study and the data are very convincing and very interesting. It makes a compelling case for the model proposed and makes many original observations and far reaching observations, such as the role of nucleo-cytoplasmic compartmentalization and P-bodies in implementing developmental decisions. Since the notion that P-bodies have a function at all has been strongly questioned in recent years, this study will be very useful for the field.
The only limitations that I have concerns the readability of the manuscript. It is extremely dense and that makes it a laborious read. Furthermore, the manuscript is not particularly well motivated, such that it is not very obvious what questions the authors are after. This becomes more or less clear only slowly as the reader progresses, or in the second read. Therefore, this very nice piece of work may escape people who are not working on fission yeast mating and meiosis, which would be a pity. I therefore recommend working on better motivating the study and its different parts for a general audience, streamlining the fission yeast intricacies and explaining more precisely what is conceptually learnt from these studies, on a broad sense and possibly in a way that would be relevant beyond the model used. This paper is opening a reach area of research and it would be unfortunate to not make that point more clearly.
Overall, this is a very carefully controlled study and the data are very convincing and very interesting. It makes a compelling case for the model proposed and makes many original observations and far reaching observations, such as the role of nucleo-cytoplasmic compartmentalization and P-bodies in implementing developmental decisions. Since the notion that P-bodies have a function at all has been strongly questioned in recent years, this study will be very useful for the field.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary
The study by Araoyinbo et al. explores the role of the RNA-binding protein Mei2 in fission yeast zygotic development. It highlights Mei2's cytosolic functions, its interaction with P-bodies, and nucleocytoplasmic shuttling. Mei2's regulation by Mei3 and Pat1, and the importance of its RNA recognition motifs (RRM1 and RRM3) are also discussed.
The main conclusion of the manuscript is somewhat unexpected from previous studies about Mei2. Particularly, the cytoplasmic function of Mei2 is a novel point in this field.
Lots of experiments have been done to make the scenario of the manuscript. The experiments and results are technically sound, and I potentially agree with the interpretation by the authors. It would require some more explanation as well as additional experiments to conclude in the way the authors wish to do.
Major points
Minor points
General assessment: strengths and limitations:
Strengths: It provides novel understanding of molecular mechanisms of meiotic initiation of fission yeast. Technically sound. Lots of experiments. Limitations: The story is very confusing and difficult to catch. Explanation can be simplified.
Advance: compare the study to existing published knowledge: does it fill a gap? What kind of advance does it make (conceptual, clinical, fundamental, methodological, incremental,,,,)? It is a big advancement. It is conceptually novel regarding how meiosis is initiated in fission yeast.
Audience: which communities will be interested/influenced, what kind of audience (broad, specialized, clinical, basic research, applied science, fields and subfields,,,) It is mainly for audience of basic research, biology, molecular mechanism of gene explanation, meiosis or yeast cellular events. For non-yeast researchers, this manuscript is probably very hard to read/understand, although the authors tried to generalize yeast-specific events with general words.
Describe your expertise:
Yeast genetics, Meiosis, Cell biology, Gene expression regulation
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
'The authors do not wish to provide a response at this time.'
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary
This manuscript presents a large-scale comparative genomics analysis of Salmonella genomes to identify and characterize the repertoire of Type VI Secretion System (T6SS) effectors. The authors combine bioinformatic predictions with experimental validation of one novel toxin domain (Tox-Act1), revealing a unique catalytic activity not previously reported in bacterial toxins. While the study is comprehensive and offers valuable insights into T6SS diversity, the insufficient description of computational methods and limited accessibility of underlying data reduce reproducibility and impact.
Major comments
Minor comments
This is a comprehensive study involving a large number of Salmonella genomes, potentially identifying many new T6SS effectors and toxic activities. One new domain analyzed in this work is experimentally investigated and shown to have a unique catalytic activity not previously observed in toxins. However, the bioinformatic methods are not described in sufficient detail, making it difficult to assess or reproduce the work. Protein accession numbers are missing, even for representative toxins, and locus tags are not traceable, making the identified effectors not readily accessible. There are many inaccuracies throughout the text and supplemental data. The Tox-Act1 domain investigated is labeled as an acyltransferase, but the evidence only supports phospholipid-degrading activity. While the study includes many graphs and histograms, they often obscure the main findings. Consequently, the audience is likely to be limited.
Nevertheless, despite these concerns, I believe this is an important work that could be valuable to the broad community once a more thorough revision is undertaken, not only by addressing the specific comments raised, but also by rechecking the analyses, reorganizing the presentation, and ensuring that all data and annotations are clearly accessible and traceable.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
The manuscript titled "Genome-directed study reveals the diversity of Salmonella T6SS effectors and identifies a novel family of lipid-targeting antibacterial toxins" presents a comprehensive in silico analysis of T6SS-associated effector and immunity genes across approximately 10,000 Salmonella genomes. In addition, the authors selected one of the newly identified effectors, Tox-Act1, for detailed biochemical characterization. To my knowledge, this study represents the most extensive genome-wide mining effort to date for T6SS-associated effectors and immunity proteins in Salmonella, employing a range of state-of-the-art computational prediction tools. The in vitro enzymatic characterization of Tox-Act1 further validates the in silico approach and adds a novel functional perspective to the dataset. Overall, the study provides a rich and comprehensive dataset. However, for readers without a strong bioinformatics background, the logic and workflow of the in silico prediction pipeline may be challenging to follow. Consequently, my comments focus primarily on the biochemical analysis of Tox-Act1, rather than the computational aspects of the study.
Major comments:
Minor Comments:
Referee cross-commenting
I agree with Reviewer #3 that the authors should provide more details on their search for better reproducibility.
This manuscript presents a large-scale in silico analysis of Salmonella T6SS effectors and immunity proteins, accompanied by the biochemical characterization of a novel phospholipase effector, Tox-Act1. The genome-wide dataset is comprehensive, representing the most extensive mining effort of its kind to date. The study is strengthened by in vitro validation of Tox-Act1 activity and its role in interbacterial competition. However, the manuscript would benefit from additional experimental data to confirm key mechanistic aspects, including T6SS-dependent secretion of Tox-Act1, its toxicity toward target cells lacking immunity, and the contribution of phospholipase activity to its antibacterial function. Comparative assays with established T6SS phospholipases (e.g., Tle1) are recommended to clarify enzymatic potency. Further, the authors should apply their phospholipase assay to test TseH activity and resolve long-standing questions in the field. Several areas also require clarification or correction, including inconsistencies in reported genome counts, incomplete figure legends, unclear terminology (e.g., "Orphan clusters"), and missing experimental controls (e.g., protein expression levels, full lipidomic dataset). Minor edits to improve clarity and consistency are also suggested. Overall, the study is significant and of high potential impact but requires additional experimental validation and revisions to improve clarity and completeness.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary: In this study, authors used in silico approaches to analyse 10,000 bacterial genomes and identified 128 candidate effectors secreted via the T6SS of Salmonella. Among these, Tox-Act1 was selected for detailed characterisation. The authors demonstrated that Tox-Act1 harbours a permuted NlpC/P60 catalytic domain with phospholipase activity, targeting key membrane lipids. Furthermore, they confirmed that Tox-Act1 is secreted in a T6SS-dependent manner and enhances bacterial competitiveness during gut colonisation in mice, providing new insights into lipid-targeting toxin domains in interbacterial interactions. My concerns raised are all minor and should be readily addressable by the authors.
Minor Concerns:
Line 279-280: The statement that the peptidoglycan is not a target of Tox-Act1 is somewhat strong at this stage of the manuscript. The preservation of cell shape does not necessarily imply that the peptidoglycan remains unaltered at a subcellular level. Given that Tox-Act1 belongs to the NlpC/P60 family, members of which include known peptidases, the authors should moderate this assertion. Replacing "is not" with "is likely not" or using conditional phrasing would be more appropriate here.
Lines 328-331: The conclusion that the Tox-Act1 clade is deployed in biological conflicts is not fully explained or substantiated. The authors are encouraged to provide a brief rationale to support this conclusion.
Figure 4D: There appears to be a labelling inconsistency. The immunity protein is referred to as "Slmm15," which may relate to the original name of Tox-Act1 (i.e., STox_15), but the correct label should likely be "Imm-Act1."
Line 401 and elsewhere: The correct spelling is "L-arabinose" with a capital "L". The manuscript should be checked for consistency in this regard.
Throughout the text and figures: Bacterial species names are often incorrectly formatted, e.g., "S. Panama" (Line 226) should be written in scientific style as S. panama, with italics and the species name in lowercase. A systematic revision of species names is recommended to enhance rigour.
Figure 3D: The X-axis labelling is somewhat confusing. The use of terms such as "attackers" and "prey" is misleading in this context, as the experiment tests the in vivo survival capacity of different Salmonella strains (WT or T6SS mutants mixed with toxin/immunity double mutants) in a mouse model, rather than a direct bacterial killing assay. Clarifying this would greatly improve readability.
Overall, this study is well-executed. The approach used to identify a previously uncharacterised diversity of T6SS effectors in Salmonella is robust and provides a valuable framework that could be extended to other systems involved in interbacterial competition. This renders the work relevant and of interest for publication. While the manuscript occasionally lacks clarity in explaining the rationale behind certain experimental choices, the narrative remains generally accessible.
Field of expertise: Secretion systems, interbacterial competition, bacterial predation, live-cell imaging, protein network
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Manuscript number: RC-2023-02191
Corresponding author: Jan Rehwinkel
The authors wish to thank all three reviewers and the Review Commons team for carefully evaluating our study. We have addressed all points raised as detailed below.
We have thoroughly revised our bulk RNAseq analysis, which is now performed at the transcript level using the latest GENCODE release. We have updated Figure 3 and associated supplementary figures and tables. This change from gene to transcript level was important for accurate motif analysis as requested by reviewer 2: matching promoters to individual IFN-regulated transcripts – rather than aggregating all promoters per gene – avoids significant signal dilution. This strategy yields higher-resolution expression data and is biologically preferable. Indeed, several well characterised IFN-regulated RNAs (e.g., the ADAR1-202 transcript encoding the p150 isoform) originate from promoters located far from the constitutive promoters of their host genes. In our revised manuscript, we now provide in the new supplementary figure 13 the requested promoter motif analysis. Using two computational approaches – de novo motif search and analysis of a curated motif database – we find strong enrichment of interferon-stimulated response elements (ISREs) in promoters of type I IFN regulated transcripts. No other motifs reached similarly high levels of enrichment, and our analysis did not reveal differences between different type I IFNs. These new data show that all type I IFNs engage a common regulatory pathway, supporting our overall conclusion that different type I IFNs do not induce qualitatively different responses in PBMCs.
Regrettably, in the process of analysing the bulk RNAseq data at transcript level, we noticed that our original lncRNA analysis contained numerous false positives. Closer inspection showed that many “differentially expressed” LNCipedia models were likely not full-length transcripts and commonly shared a single IFN-induced set of exons that artificially inflated expression estimates for every overlapping model. To correct this issue, we replaced LNCipedia with the latest high-quality non-coding RNA catalogue from GENCODE, most entries of which were defined by full-length RNA sequencing [1]. We also tightened our filtering criteria and now report only transcripts that are robustly expressed in our dataset and are either classified as high-confidence by GENCODE or robustly supported at every splice junction by our RNAseq.
We hope our manuscript is sufficiently improved and suitable for publication in PLoS Biology. New or revised text is highlighted in green in our revised manuscript.
Reviewer #1
Evidence, reproducibility and clarity:
The study can be directly connected to a landmark paper in the field (Mostafavi et al. , Cell 2016). By comparison with this study, the authors use improved technologies to address the question if and how responses to type I IFN differ between human peripheral blood-derived cells types. In line with Mostafavi et al. the authors conclude that only a comparably low number of interferon-stimulated genes (ISG) is induced in all cell types and that considerable differences exist between cell types in the IFN-induced transcriptome. The authors address a second relevant aspect, whether and how the many different subtypes of type I IFN differ in the way they engage IFN signals to produce transcriptome changes. The data lead the authors to conclude that any differences are of quantitative rather than qualitative nature.
The authors' conclusions are based on a mass cytometry approach to phenotype STAT activation in different cell types, bulk RNA sequencing to study ISG expression in PBMC, and single cell sequencing to study ISG responses in individual cell types. The data are solid, clear and reproducible in biological replicates (eg different blood donors).
Significance: While some of the data can be considered confirmatory, the comprehensive analysis of cell-type specificity and IFN-I subtype specificity advances the field and provides a reference for future analyses. The study is complete and there is no obvious lack of a critical experiment. The number of scientists interested in the multitude of open questions around type I IFN is large, thus the study is likely to attract a broad readership.
We thank the reviewer for her/his positive assessment of our study.
The biggest limitation is to my opinion the low sequencing depth of scRNAseq which is clearly the downside of this technology. Using 11 hematopoietic cell types and bulk RNA sequencing the total number of ISG was determined to be 975 by Mostafavi et al. and the core ISG numbered 166. This is in stark contrast to this studies' 10 core ISG. The authors limitations paragraph should discuss the fact that scRNAseq reduces the overall ISG number that can be analyzed.
Thank you for this valid comment. We amended the limitations paragraph as requested. We agree that the Mostafavi et al. 2016 Cell paper [2] is important but note that there are many differences to our study: Mostafavi et al. use mice, a seemingly very high IFN dose (10,000 Units) and microarrays (not RNAseq).
A minor point concerns the 25 supplementary figures of the study. There must be a better way to support the conclusions with the necessary data.
We agree that our supplementary materials are extensive. However, this is not unusual for studies reporting multiple large datasets. We would be delighted to organise our supplementary information differently in due course according to journal guidelines.
Reviewer #2
Evidence, reproducibility and clarity:
The manuscript entitled “Single-cell analysis of signalling and transcriptional responses to type I interferon" by Rigby et al. examines the response to type I IFN subtypes in PBMCs using an integrative proteomics and transcriptomics approach. Some of the analysis could be deepened to provide better insights into what governs the magnitude of change in gene expression as well as the cell type-specific response to expression and generate more excitement for the study.
We thank the reviewer for evaluating our study and the suggestions made.
*Major Comments: *
Next, we conducted a complementary analysis using known transcription factor (TF) motifs from the JASPAR database [4]. We screened all promoters of annotated RNAs using clustered JASPAR motifs and Z-standardised motif scores relative to all high-confidence GENCODE RNAs, including those not expressed in PBMCs. We reasoned that TFs actively mediating IFN responses would likely bind promoters with high motif scores (Z ≥ 2), while promoters with low scores (Z ≤ -1) would represent an unregulated background. This approach produced two sets of RNAs per TF cluster: putatively regulated and unregulated. We then restricted each set to RNAs expressed in our dataset and associated each transcript with its estimated fold change in response to each type I IFN, regardless of statistical significance. Next, we compared median fold changes between the likely regulated and unregulated sets across all TF clusters and IFN subtypes (Figure S13b). Among all tested TF motifs, only the ISRE-like cluster showed strong and consistent associations with transcriptional changes across all IFN subtypes. We also observed statistically significant but much weaker associations for other TFs, including a known negative regulator of innate antiviral signaling, NRF1 [5]. However, effect sizes for these motifs were dwarfed by those of ISRE-like motifs, suggesting that no JASPAR TFs other than those within the ISRE-like cluster play a major role in PBMCs under our conditions. Overall, these findings support the idea that all type I IFNs engage a common regulatory pathway, differing primarily in the magnitude rather than the nature of their transcriptional effects.
How do they relate to the activation of kinases by IFN subtypes?
We did not analyse the activation of the canonical kinases (i.e., TYK2 and JAK1) downstream of IFNAR. This would be interesting and may be possible using phospho-specific antibodies to these kinases in our CyTOF setup. However, this would require a very large investment of time and resources to identify specific antibodies, optimise a new CyTOF staining panel and to acquire and analyse new datasets. We therefore believe this should be pursued as a separate future study.
*Are there distinct features that dictate differential responses in monocytes and lymphocytes? *
Following the computational approach described above, we applied STREME to identify DNA motifs that could distinguish promoters associated with monocyte- and lymphocyte-specific ISGs. Regrettably, this analysis did not yield any significant motifs, likely due in part to the limited number of genes in each category.
Thank you for this suggestion. We tried using the same scale for all heatmaps. However, given that the values for pSTAT1 are higher than those for other pSTATs, the resulting heatmaps did not show differences for the other pSTATs well. We therefore decided to leave these panels unchanged. Please also note that Figures 2b and S3b provide comparison between pSTATs (and other markers) using the same scale.
Minor Comments:
The title of subsections are a bit generic (e.g "Analysis of the signalling response to type I IFNs using mass cytometry". Consider updating them to reflect some of the findings from each analysis.* Thank you for this suggestion. We have amended sub-headers accordingly.
Figure 3 and S3 - Increase the heatmap scale to better appreciate changes in gene expression.*
The scales have been enlarged for better visibility as requested.
Thank you for the suggestion. We combined these panels.
Figure 5 and several accompanying supplementary figures already depict ISGs unique to IFN subtypes or cell types. Whilst we appreciate the suggestion, we prefer not to add additional figures to avoid redundancies.
Thank you for this comment. We changed the presentation of the GO analysis in Fig S11 by sorting on p-value (instead of % of hits in category). We hope this shows more clearly that GO category enrichment amongst genes encoding IFN-induced transcripts had high statistical significance (log10 p-values of about -5 or lower for many categories).
Significance:* ** The authors provide an extensive compendium of cell type specific changes in response to type I IFN stimulation. They have created a public repository which extends the value of this dataset. *
Audience: *** This is a valuable resource for immunologists, virologists, and bioinformaticians.*
Thank you for these encouraging comments.
Reviewer #3
Evidence, reproducibility and clarity:
*Summary *
Rigby and collaborators analyzed the signaling responses and changes in gene expression of human PBMCs stimulated with different IFN type I subtypes, using mass cytometry, bulk and single-cell RNA sequencing. Their study represents the first single-cell atlas of human PBMCs stimulated with five type I IFN subtypes. The generated datasets are useful resources for anyone interested in innate immunity. The data and the methods are well presented. We thus recommend publication.
Thank you for your positive assessment of our work and for recommending publication.
*Major comments: *
*Two of the key conclusions are not very convincing. *
First, the authors claim that the magnitude of the responses varied between the 5 types of IFNs, however, as they point out in the 'limitation' paragraph, doses of the different IFNs were normalized using bioactivity. Knowing that this bioactivity is based on assays performed on A549 lung cells, this normalization likely induces a bias. How do the authors explain similar antiviral bioactivity but differing magnitudes of modulation of ISG expression? Would the authors expect the same differences of expression between the several IFNs tested in A549 cells? We thus recommend being very cautious when comparing magnitude of the response between the 5 types of IFNs.
We thank the reviewer for this important point and included the following reasoning in our discussion:
“An important technical consideration for our study was the normalisation of type I IFN doses used to treat cells (see also ‘Limitations of the study’ below). We relied on bioactivity (U/ml) that is measured by the manufacturer of recombinant type I IFNs using a cytopathic effect (CPE) inhibition assay. In brief, the lung cancer cell line A549 is treated with type I IFN and is infected with the cytopathic encephalomyocarditis virus (EMCV). Control cells not treated with IFN are killed by EMCV, whereas cells treated with sufficient IFN survive. How, then, is it possible that different type I IFNs induce differing magnitudes of STAT phosphorylation and ISG expression despite being used at the same bioactivity? Cell survival in the CPE inhibition assay may be due to one or a few ISGs. Indeed, single ISGs can mediate powerful antiviral defence. For example, MX1 is crucial for host defence against influenza A virus [6]. Thus, similar bioactivity of different IFNs in A549 cells against EMCV-triggered cell death may not reflect the breadth of effects on many ISGs. Moreover, IFN-induced survival of A549 cells following EMCV infection is a binary readout. Induction of the relevant ISG(s) mediating protection beyond a threshold required for cell survival is unlikely to register in this assay. Thus, similar antiviral bioactivity (in the CPE inhibition assay) and differing magnitudes of modulation of ISG expression (at transcriptome level) are compatible.”
We believe inclusion of this paragraph demonstrates an appropriate level of caution in our data interpretation. Further, we would expect to make similar observations if we were to apply transcriptomic analysis to A549 cells treated with different type I IFNs. However, given our focus in this study on primary, normal cells, we decided not to pursue work with the transformed and lab adapted A549 cell line.
Second, the qualitatively different responses to type I IFN subtypes claimed by the authors were not apparent. This seems true at the level of the bulk population (Fig. S10) but not at cell-type level (Fig. S15/S16).
We believe there may be a misunderstanding here. In relation to Figure S10, we do not claim “qualitatively different responses to type I IFN subtypes”. Instead, we conclude that “differences in expression between the different type I IFNs were quantitative” (page 8; lines 229-230, now: 238-239). Moreover, Figures S15/S16 (now: S16/S17) do not refer to analyses of responses to different type I IFN subtypes.
The authors state (line 311-312) that 'Consistent with our bulk RNAseq data, differences were again quantitative rather than qualitative' at the cell-type level. The response between cell types seems very different to us since a core set of only 10 ISGs are shared by all cell types and all 5 type I IFNs. Knowing that the expression of hundreds, sometimes thousands of genes, are induced by IFN, this seems like a rather small overlap (and thus qualitatively different responses). Fig S15 and S16 nicely illustrate that the responses are qualitatively different between cell-type. Please modify this conclusion accordingly.
Thank you for highlighting this. The statement in lines 311-312 does not refer to differences between cell types but to differences between type I IFN subtypes. We are sorry this was not clear and changed this sentence (now lines 357-358). Furthermore, we have made it clearer in the revised text that qualitative differences were observed between cell types (e.g. lines 329 and 350-352).
*No additional experiments are needed to support the claims. However, we believe that two additional analyses could provide useful information. *
The levels of IFNAR1 and IFNAR2 expressed at the plasma membrane probably vary between cell types and may thus influence the magnitude of the IFN response. While it would be difficult to measure these levels by flow cytometric analysis on the different cell types, could the authors extract information from their scRNAseq analysis on the expression level of IFNAR1/2 in all cell types? This would give a hint about potential differences in expression (and thus in magnitude).
We analysed IFNAR1/2 transcript levels in our scRNAseq dataset (Figure R1 below). Unfortunately, for many cells, IFNAR1 and IFNAR2 transcripts were not detected (see width of violin plots at zero), probably due to low sequencing depth inherent to scRNAseq analysis. We therefore prefer not to draw conclusions from these data.
Could the authors investigate further the expression of lncRNAs at the single-cell levels? It would be useful to also define a core set of lncRNAs that are shared between cell types and IFN subtypes. If such a core set does not exist (since lncRNAs are less conserved than coding genes), it would be nice to mention it.
Thank you for this suggestion. The expression of lncRNAs is generally lower than protein-coding genes, resulting in high drop-out rates in 10X datasets. Indeed, Zhao et al. comment that “current development of single-cell technologies may not yet be optimized for lncRNA detection and quantification” [7]. We only detected a small number of lncRNAs in our scRNAseq analysis, and only four lncRNAs were significantly differentially expressed between cell types. We thus could not perform a meaningful analysis of lncRNAs in our scRNAseq dataset. This is now mentioned in the limitations paragraph at the end of the manuscript.
Minor comments:
There is a typo in line 355 Fig.4C =>6C.
Thank you for spotting this.
***Referees cross-commenting** *
We agree with Reviewer 1 that the low sequencing depth of scRNAseq restricts the analysis and must be discussed in the 'limitation' paragraph. This would explain why the authors identified only 10 ISGs that are common to all cell types and all 5 IFN subtypes. Of note, as a comparison, Shaw et al (10.1371/journal.pbio.2004086) identified a core set of 90 ISGs that are upregulated upon IFN treatment in cells isolated mainly from kidney and skin of nine mammalian species ("core mammalian ISGs"). It is thus expected that stimulated blood cells isolated from a single mammalian species share more than 10 ISGs.
We amended the limitations section as requested. Shaw et al. [8] used a single type I IFN (universal or IFNα, depending on species) at a very high dose (1000 U/ml). Taken together with the use of bulk RNAseq in this study, it is unsurprising that our work identified fewer core ISGs. We believe our small list of core ISGs is nonetheless both a high confidence and a high utility set of ISGs: these genes are induced by multiple type I IFNs, in all major cell types in blood and their regulation can be measured even when sequencing depth is low.
Significance (Required)
*Multiple single-cell RNAseq analysis of PBMCs, stimulated or not, have been previously performed in multiple contexts (for instance with PBMCs isolated from the blood of patients infected with influenza virus or SARS-CoV-2). The technical advance is thus limited. *
*However, the work represents a conceptual advance for the field since it provides the first single-cell atlas of PBMCs stimulated with five type-I IFN subtypes. The generated datasets represent a great resource for anyone interested in innate immunity (virologists, immunologists and cancerologists). *
Of note, we are studying innate immunity in the context of RNA virus infection but we have no expertise on scRNA sequencing. We may thus have missed a flaw in the analyses.
We thank the reviewer for their positive assessment of the advances of our study and the value of our IFN resource.
A
B
C
D
Figure R1. IFNAR1/2 expression in scRNAseq data.
Violin plots showing expression of IFNAR1 (A,C) or IFNAR2 (B,D) in different cell types. In (A,B), data were pooled across conditions. In (C,D), data are shown separately for unstimulated control cells and cells stimulated with different type I IFNs.
References
Kaur G, Perteghella T, Carbonell-Sala S, Gonzalez-Martinez J, Hunt T, Madry T, et al. GENCODE: massively expanding the lncRNA catalog through capture long-read RNA sequencing. bioRxiv. 2024. Epub 20241031. doi: 10.1101/2024.10.29.620654. PubMed PMID: 39554180; PubMed Central PMCID: PMCPMC11565817. Mostafavi S, Yoshida H, Moodley D, LeBoite H, Rothamel K, Raj T, et al. Parsing the Interferon Transcriptional Network and Its Disease Associations. Cell. 2016;164(3):564-78. Epub 2016/01/30. doi: 10.1016/j.cell.2015.12.032. PubMed PMID: 26824662; PubMed Central PMCID: PMCPMC4743492. Bailey TL. STREME: accurate and versatile sequence motif discovery. Bioinformatics. 2021;37(18):2834-40. doi: 10.1093/bioinformatics/btab203. PubMed PMID: 33760053; PubMed Central PMCID: PMCPMC8479671. Rauluseviciute I, Riudavets-Puig R, Blanc-Mathieu R, Castro-Mondragon JA, Ferenc K, Kumar V, et al. JASPAR 2024: 20th anniversary of the open-access database of transcription factor binding profiles. Nucleic acids research. 2024;52(D1):D174-D82. doi: 10.1093/nar/gkad1059. PubMed PMID: 37962376; PubMed Central PMCID: PMCPMC10767809. Zhao T, Zhang J, Lei H, Meng Y, Cheng H, Zhao Y, et al. NRF1-mediated mitochondrial biogenesis antagonizes innate antiviral immunity. The EMBO journal. 2023;42(16):e113258. Epub 20230706. doi: 10.15252/embj.2022113258. PubMed PMID: 37409632; PubMed Central PMCID: PMCPMC10425878. Grimm D, Staeheli P, Hufbauer M, Koerner I, Martinez-Sobrido L, Solorzano A, et al. Replication fitness determines high virulence of influenza A virus in mice carrying functional Mx1 resistance gene. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(16):6806-11. Epub 20070410. doi: 10.1073/pnas.0701849104. PubMed PMID: 17426143; PubMed Central PMCID: PMCPMC1871866. Zhao X, Lan Y, Chen D. Exploring long non-coding RNA networks from single cell omics data. Comput Struct Biotechnol J. 2022;20:4381-9. Epub 20220804. doi: 10.1016/j.csbj.2022.08.003. PubMed PMID: 36051880; PubMed Central PMCID: PMCPMC9403499. Shaw AE, Hughes J, Gu Q, Behdenna A, Singer JB, Dennis T, et al. Fundamental properties of the mammalian innate immune system revealed by multispecies comparison of type I interferon responses. PLoS Biol. 2017;15(12):e2004086. Epub 2017/12/19. doi: 10.1371/journal.pbio.2004086. PubMed PMID: 29253856.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary
Rigby and collaborators analyzed the signaling responses and changes in gene expression of human PBMCs stimulated with different IFN type I subtypes, using mass cytometry, bulk and single-cell RNA sequencing. Their study represents the first single-cell atlas of human PBMCs stimulated with five type I IFN subtypes. The generated datasets are useful resources for anyone interested in innate immunity. The data and the methods are well presented. We thus recommend publication.
Major comments:
Two of the key conclusions are not very convincing.
First, the authors claim that the magnitude of the responses varied between the 5 types of IFNs, however, as they point out in the 'limitation' paragraph, doses of the different IFNs were normalized using bioactivity. Knowing that this bioactivity is based on assays performed on A549 lung cells, this normalization likely induces a bias. How do the authors explain similar antiviral bioactivity but differing magnitudes of modulation of ISG expression? Would the authors expect the same differences of expression between the several IFNs tested in A549 cells? We thus recommend being very cautious when comparing magnitude of the response between the 5 types of IFNs.
Second, the qualitatively different responses to type I IFN subtypes claimed by the authors were not apparent. This seems true at the level of the bulk population (Fig. S10) but not at cell-type level (Fig. S15/S16). The authors state (line 311-312) that 'Consistent with our bulk RNAseq data, differences were again quantitative rather than qualitative' at the cell-type level. The response between cell types seems very different to us since a core set of only 10 ISGs are shared by all cell types and all 5 type I IFNs. Knowing that the expression of hundreds, sometimes thousands of genes, are induced by IFN, this seems like a rather small overlap (and thus qualitatively different responses). Fig S15 and S16 nicely illustrate that the responses are qualitatively different between cell-type. Please modify this conclusion accordingly.
No additional experiments are needed to support the claims. However, we believe that two additional analyses could provide useful information.
The levels of IFNAR1 and IFNAR2 expressed at the plasma membrane probably vary between cell types and may thus influence the magnitude of the IFN response. While it would be difficult to measure these levels by flow cytometric analysis on the different cell types, could the authors extract information from their scRNAseq analysis on the expression level of IFNAR1/2 in all cell types? This would give a hint about potential differences in expression (and thus in magnitude).
Could the authors investigate further the expression of lncRNAs at the single-cell levels? It would be useful to also define a core set of lncRNAs that are shared between cell types and IFN subtypes. If such a core set does not exist (since lncRNAs are less conserved than coding genes), it would be nice to mention it.
Minor comments:
There is a typo in line 355 Fig.4C =>6C.
Referees cross-commenting
We agree with Reviewer 1 that the low sequencing depth of scRNAseq restricts the analysis and must be discussed in the 'limitation' paragraph. This would explain why the authors identified only 10 ISGs that are common to all cell types and all 5 IFN subtypes. Of note, as a comparison, Shaw et al (10.1371/journal.pbio.2004086) identified a core set of 90 ISGs that are upregulated upon IFN treatment in cells isolated mainly from kidney and skin of nine mammalian species ("core mammalian ISGs"). It is thus expected that stimulated blood cells isolated from a single mammalian species share more than 10 ISGs.
Multiple single-cell RNAseq analysis of PBMCs, stimulated or not, have been previously performed in multiple contexts (for instance with PBMCs isolated from the blood of patients infected with influenza virus or SARS-CoV-2). The technical advance is thus limited.
However, the work represents a conceptual advance for the field since it provides the first single-cell atlas of PBMCs stimulated with five type-I IFN subtypes. The generated datasets represent a great resource for anyone interested in innate immunity (virologists, immunologists and cancerologists).
Of note, we are studying innate immunity in the context of RNA virus infection but we have no expertise on scRNA sequencing. We may thus have missed a flaw in the analyses.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript entitled "Single-cell analysis of signalling and transcriptional responses to type I interferon" by Rigby et al. examines the response to type I IFN subtypes in PBMCs using an integrative proteomics and transcriptomics approach. Some of the analysis could be deepened to provide better insights into what governs the magnitude of change in gene expression as well as the cell type-specific response to expression and generate more excitement for the study.
Major Comments:
Minor Comments:
Significance:
The authors provide an extensive compendium of cell type specific changes in response to type I IFN stimulation. They have created a public repository which extends the value of this dataset.
Audience:
This is a valuable resource for immunologists, virologists, and bioinformaticians.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The study can be directly connected to a landmark paper in the field (Mostafavi et al. , Cell 2016). By comparison with this study, the authors use improved technologies to address the question if and how responses to type I IFN differ between human peripheral blood-derived cells types. In line with Mostafavi et al. the authors conclude that only a comparably low number of interferon-stimulated genes (ISG) is induced in all cell types and that considerable differences exist between cell types in the IFN-induced transcriptome. The authors address a second relevant aspect, whether and how the many different subtypes of type I IFN differ in the way they engage IFN signals to produce transcriptome changes. The data lead the authors to conclude that any differences are of quantitative rather than qualitative nature. The authors' conclusions are based on a mass cytometry approach to phenotype STAT activation in different cell types, bulk RNA sequencing to study ISG expression in PBMC, and single cell sequencing to study ISG responses in individual cell types. The data are solid, clear and reproducible in biological replicates (eg different blood donors).
While some of the data can be considered confirmatory, the comprehensive analysis of cell-type specificity and IFN-I subtype specificity advances the field and provides a reference for future analyses. The study is complete and there is no obvious lack of a critical experiment. The number of scientists interested in the multitude of open questions around type I IFN is large, thus the study is likely to attract a broad readership.
The biggest limitation is to my opinion the low sequencing depth of scRNAseq which is clearly the downside of this technology. Using 11 hematopoietic cell types and bulk RNA sequencing the total number of ISG was determined to be 975 by Mostafavi et al. and the core ISG numbered 166. This is in stark contrast to this studies' 10 core ISG. The authors limitations paragraph should discuss the fact that scRNAseq reduces the overall ISG number that can be analyzed.
A minor point concerns the 25 supplementary figures of the study. There must be a better way to support the conclusions with the necessary data.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
We would like to thank all reviewers assigned by Review Commons for their thoughtful and constructive feedback, which helped us to further improve the quality and clarity of our manuscript. In this study, we developed a novel fluorescence-based live-cell imaging platform for detecting mitochondria-endoplasmic reticulum contact sites (MERCS), which we named MERCdRED. This system enables quantitative analysis of MERCS dynamics in living cells by combining stable gene expression of dimerization-dependent fluorescent proteins with single-cell cloning. Using this tool, we uncovered a nutrient-dependent regulatory mechanism of MERCS formation mediated by the ER-localized tethering protein PDZD8. We appreciate that all the reviewers acknowledged the methodological robustness of this work. In response to reviewers' comments, we will significantly improve the manuscript by adding the live-cell imaging to assess the reversible propertyof MERCdRED, and investigating the physiological impacts of MERCS remodeling in regulating metabolism in response to nutrient starvation. We believe that both the methodological advance and the biological findings presented in this study will be of broad interest to the cell biology community.
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
Summary: In this study, the authors successfully established a stable cell line expressing MERCdRED, a dimerization-dependent fluorescent protein (ddFP)-based sensor for monitoring mitochondrial-ER contact sites (MERCS). Through light and electron microscopy analyses, they demonstrated that MERCS formation is regulated by nutrient availability and requires PDZD8. While the work is technically sound and well-presented, the biological implications of nutrient-dependent MERCS regulation remain underexplored.
Major Concerns: Although the manuscript is methodologically robust and suitable for a Methods-type article, its biological significance is limited. The findings primarily serve as proof-of-concept for the MERCdRED tool, without substantially advancing our understanding of MERCS regulation.
We appreciate the reviewer for acknowledging the methodological robustness of our study. We would like to respectfully emphasize that, using the MERCdRED cell system, we uncovered the distinct features of MERCS dynamics by comparing structures of various sizes (Figure 4A-D). Furthermore, we discovered an unexpected biological finding: nutrient starvation leads to a reduction in MERCS formation, which contrasts with previous reports using cell lines (former Figure 4E-H). Additionally, we revealed that PDZD8 mediates nutrient-dependent MERCS regulation (former Figure 4E-H).
To clarify these findings, we have now separated the former Figure 4 into two distinct figures (now Figure 4 and 5). Furthermore, to assess the functional relevance of PDZD8-mediated MERCS regulation upon nutritional change, we will perform rescue experiments by overexpressing PDZD8 in starved cells, along with a metabolomic analysis in these conditions. We will add these new data in Figure 6.
Taken together, we believe that our data provide novel mechanistic insights into how MERCS are modulated and utilized for the regulation of metabolism under physiological stress, thereby contributing to a deeper understanding of the roles and regulation of MERCS beyond the scope of a mere proof-of-concept study.
Reviewer #1 (Significance (Required)):
To enhance the impact of the study, the authors could use this sensor to investigate novel biological questions-such as the molecular pathways linking nutrient sensing to MERCS dynamics-or explore downstream activities of nutrient-dependent MERCS formation. Deeper mechanistic insights would significantly strengthen the work's contribution to the field.
We thank the reviewer for their constructive suggestions. We fully agree that the MERCdRED cell system has great potential for investigating upstream signaling pathways regulating MERCS dynamics, as well as the downstream consequences of nutrient-dependent MERCS modulation. As mentioned above, this study already presents important findings, including the discovery of PDZD8 as a key protein linking the nutrient starvation and MERCS remodeling, and a relationship between MERCS dynamics and contact site size.
To further assess the biological consequence of the MERCS remodeling, we will perform metabolomics analysis in PDZD8-overexpressing cells under starved conditions.
Additionally, to further reinforce the utility of MERCdRED and extend the findings presented in this study, we performed live-cell imaging experiments using MERCdRED. The preliminary results demonstrated dynamic and reversible changes in MERCS in response to nutrient starvation and subsequent recovery (Please see the response to Reviewer 3 below, Reviewer-only Figure 1).
These new data will significantly strengthen the contribution of this study to the field.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
This manuscript entitled "Live-cell imaging reveals nutrient-dependent dynamics of ER-mitochondria contact formation via PDZD8" by Saeko Aoyama-Ishiwatari et al., describes a novel methodology for visualizing contacts between mitochondria and the endoplasmic reticulum (MERCs) by fluorescence microscopy. Inter-organelle contacts, defined as membrane proximities below ~30 nm, fall below the diffraction limit of conventional light microscopy. The method developed by Hirabayashi's laboratory leverages dimerization-dependent fluorescence complementation to create a reporter capable of both visualizing and quantifying ER-mitochondria contacts (MERCs).
Reviewer #2 (Significance (Required)):
This timely study provides a valuable and innovative approach to overcoming a longstanding technical limitation in the field, enabling dynamic analysis of ER-mitochondria contacts.
We appreciate the reviewer for recognizing the timeliness and innovation of our work.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
In this manuscript, the authors develop a new system to study mitochondrial-ER contact sites in living mouse embryonic fibroblast cells and explore the impact that nutritent starvation has on these contact sites in real time. By stably expressing a bicistroic reporter construct of a dimerization-dependent fluorescent protein that will generate a signal once the two moities, one anchored in the ER by Sec61b, and the other anchored in the outer membrane of mitochondria, via TOM20, comme in close apposition. This cell model is validated using sofisticated CLEM experiments and via the ablation of known regulators of MERCs, such as PDZD8 and FKBP8.
Major comments
The authors claim to have developed a new for the study of MERCs. They indeed have benchmarked this system using very sophisticated CLEM approaches and through the ablation of known regulators of MERCs, all of which is very carefully performed and convincing.
We appreciate the reviewer for acknowledging our efforts in the development and validation of the MERCdRED system presented in this study.
They argue that the generation of a stable cell line via lentiviral delivery is an improvement over the transient transfection approaches that have been applied in the past (see cited references), which I would generally agree. However, they have not contrasted or compared their system to the widly-used SLPICs system from the Tito Cali group (Vallese, F. et al. An expanded palette of improved SPLICS reporters detects multiple organelle contacts in vitro and in vivo. Nat. Commun. 11, 6069 (2020)) which measures bi and tri-partite interactions with other membrane contacts, including mitochondria and ER at two specific distances, which in my opinion has been more extensivley used to study cell and tissue physiology. They accurately point out that the reversability of this and other systems is challenging and it would be important to define highlight whether the current system allows the study of reversible MERCs. It does not appear as though the reversability of MERCs has been explored in this study.
We thank the reviewer for these thoughtful suggestions and agree that further investigation into the reversibility of MERCS using the MERCdRED system would be valuable. Following the reviewer's suggestion, we performed a live-cell imaging experiment using MERCdRED to monitor dynamic changes in MERCS in response to nutrient starvation and subsequent recovery. The preliminary results were obtained as shown in Reviewer-only Figure 1, which strongly suggests the utility of MERCdRED for detecting reversible MERCS formation. The data will be added in Figure 5 if the reproducibility is confirmed. This new data set highlights the distinct utility of the MERCdRED system in studying MERCS dynamics.
We acknowledge that the SPLICS system has been widely adopted for studying membrane contact sites. In the revised manuscript, we will include a comparative discussion of MERCdRED, SPLICS, and other existing MERCS reporters, particularly with respect to their capabilities in capturing the reversible nature of these contacts.
The genetic (PDZD8, FKBP8) and nutritional (starvation) interventions are very helpful to benchmark the system. The description of the methods and data appear to be reproducible and the stastical analyses are acceptable.
We thank the reviewer for their positive evaluation of our data and analyses.
Minor comments
As mentioned above, it would be helpful to reference and compare the current study in the context of reversability, which the current MERCdRED system has the potential to provide beyond the state-of-the-art.
We thank the reviewer for this helpful suggestion. We will include additional discussion comparing the reversibility of the MERCdRED system with that of existing tools, highlighting the potential advantages of MERCdRED in capturing dynamic and reversible MERCS.
Reviewer #3 (Significance (Required)):
Significance
The major strength of this study is the development of a stable cell line that allows for the study of MERCs, which has the potential to study the reversible nature of these membrane contact sites. It is debatable as a stable cell line rather than a transient transfection offers a major advancement, even if it does make the study of the system more straightforward, especially if the phenomenon of reversibilty is to be explored.I believe that the CLEM study offers a very informative and precise way to benchmark the ddFP system. Defining how MERC formation and separation (once again the reversibility discovery) have impacts in cell physiology beyond the distances altered by starvation would improve the study. Examining the impact on calcium homeostasis, lipid metabolism, and other aspects of biology that are known to be influenced by MERCs would be interesting. As such, there are no new conceptual, mechanistic, or functional advances, simply minor technical advances in the creation of a stable cell line followed by very solid benchmarking experiments. More complex tri-partite interactions, studied elsewhere, which are conceptually very important for cell and organelle biology, have not been attempted here. Similarly, the notion of studied different types of MERCs, which have been proposed to be important for cell biology, has not been explored using this single reporter. The target audience for this study is one that is interested in membrane contact sites and quantitative biology. My expertise is in mitochondrial fluorescence imaging and biology. I am not an expert in CLEM.
We thank the reviewer for their thoughtful and detailed comments. We would like to respectfully emphasize that the establishment of a clonal cell line has enabled us to uncover a striking and unexpected biological finding-namely, that nutrient starvation leads to a reduction, rather than an increase, in MERCS formation, and that this change is regulated by PDZD8. This observation directly contradicts previous reports and highlights the value of our robust and quantitative system for re-evaluating previously held assumptions.
We agree that demonstrating the reversibility of MERCS formation using our system would further strengthen the utility and reliability of the MERCdRED platform. To address this, as mentioned above, we performed a live-cell imaging to assess the dynamic reversibility of MERCS formation (Reviewer-only Figure 1) and will add the results in the revised manuscript.
We agree that investigation of tri-partite interactions is conceptually important for understanding the broader landscape of organelle communication. However, assessing tri-partite organelle contacts is beyond the scope of this study. We recognize that this is one of the key directions for future studies and believe that the MERCdRED platform is a promising tool for exploring such complex interactions.
Regarding different types of MERCS, we would like to clarify that our study does address this point to some extent. We identified distinct features of MERCS behavior by comparing structures of different sizes-an aspect that, to our knowledge, has not been previously examined. These findings contribute conceptually to our understanding of the dynamic and heterogeneous nature of ER-mitochondria contacts.
We believe that our methodological development provides important mechanistic insights into MERCS dynamics, as described above. In line with the reviewer's suggestion, we will investigate the physiological impacts of MERCS remodeling in regulating metabolism in response to nutrient starvation. We hope these forthcoming data will further enhance the biological relevance of our findings.
Taken together, we believe our study provides both a solid technical advance and novel mechanistic insights into MERCS biology, which will be of interest to researchers working on membrane contact sites, organelle dynamics, and cell physiology.
We will revise the manuscript to more clearly convey the significance and implications of this study.
Reviewer #2
Major points:
In Figure 1E (and the rest of the manuscript), the meaning of the label "MERCdRED on Mito" is unclear. A portion of the MERCdRED signal does not co-localize with mitochondria. The authors should clearly define what "MERCdRED on Mito" represents which appears to be the intensity of the MERCdRED signal within the mitochondrial mask. How about the global MERCdRED signal intensity? When the authors knocked-out PDZD8, did the global fluorescence intensity of the MERCdRED signal decrease?
As the reviewer pointed out, some red signals appear outside of mitochondria in MERCdRED cells, which are presumably due to autofluorescence. While the global red channel fluorescence intensity also decreased upon PDZD8 conditional knockout (cKO), as shown in Reviewer-only Figure 2A, the reduction was less pronounced than the decrease observed when only the red signals on mitochondria were measured (Reviewer-only Figure 2B). We consider the mitochondrial red signals to represent MERCdRED signals, and we agree that the label "MERCdRED on Mito" may be misleading. To improve clarity, we revised the figure labels as follows: "MERCdRED" was changed to "Red channel," and "MERCdRED on Mito" was changed to "MERCdRED (Red signals on Mito)."
- While the authors demonstrate that MERCdRED can quantify a reduction in MERCs (e.g., in PDZD8 knockout conditions), it would be valuable to assess its sensitivity to increases in MERCs as well. For example, previous work from the authors (Nakamura et al., 2025) showed that FKBP8 overexpression leads to an increase in MERCs.
We thank the reviewer for suggesting this valuable experiment. To assess whether the dynamic range of MERCdRED covers increased MERCS formation, we overexpressed PDZD8 in MERCdRED cells. Notably, PDZD8 overexpression resulted in a significant increase in MERCdRED signal intensity, demonstrating that the system is indeed capable of detecting enhanced MERCS formation. These new data were added in the revised manuscript as new Figure 3D-E.
Minor points: 1. Please revise the sentence "First, signals from MERCdRED fluorescence overlapped with the mitochondrial marker Tomm20-iRFP were detected by confocal microscopy in living cells."
We revised this sentence to "First, fluorescence from MERCdRED and the mitochondrial marker Tomm20-iRFP wasdetected by confocal microscopy in living cells."
Reviewer #2
Major points: 1. The authors claim that their construct enables balanced expression of the RA and GB moieties of the reporter. This should be substantiated by showing protein expression levels via Western blot analysis.
We thank the reviewer for pointing this out. In our system, Tomm20-GB and RA-Sec61β are expressed from a single plasmid using a self-cleaving P2A peptide sequence, which ensures that the two proteins are produced in equimolar amounts upon translation. Therefore, their expression levels are expected to be approximately equal. Given that comparing the expression levels of these two proteins by Western blotting would require extensive work, including obtaining reconstituted proteins to normalize band intensities, but remains inconclusive due to the semi-quantitative nature of the method, we have decided not to pursue this approach.
Minor points:
- In Figure 2, the ER structures are not segmented in the EM images. It would enhance the manuscript to show the three-dimensional spatial relationship between mitochondria and the ER, rather than only highlighting the regions identified as contacts.
We agree that visualizing the entire ER structure would enhance the reader's understanding of the three-dimensional spatial relationship between mitochondria and the ER. However, complete segmentation of the ER in EM images is extremely labor-intensive. Given the scope and focus of this study, we have decided not to include full ER segmentation in this manuscript.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this manuscript, the authors develop a new system to study mitochondrial-ER contact sites in living mouse embryonic fibroblast cells and explore the impact that nutritent starvation has on these contact sites in real time. By stably expressing a bicistroic reporter construct of a dimerization-dependent fluorescent protein that will generate a signal once the two moities, one anchored in the ER by Sec61b, and the other anchored in the outer membrane of mitochondria, via TOM20, comme in close apposition. This cell model is validated using sofisticated CLEM experiments and via the ablation of known regulators of MERCs, such as PDZD8 and FKBP8.
Major comments
The authors claim to have developed a new for the study of MERCs. They indeed have benchmarked this system using very sophisticated CLEM approaches and through the ablation of known regulators of MERCs, all of which is very carefully performed and convincing. They argue that the generation of a stable cell line via lentiviral delivery is an improvement over the transient transfection approaches that have been applied in the past (see cited references), which I would generally agree. However, they have not contrasted or compared their system to the widly-used SLPICs system from the Tito Cali group (Vallese, F. et al. An expanded palette of improved SPLICS reporters detects multiple organelle contacts in vitro and in vivo. Nat. Commun. 11, 6069 (2020)) which measures bi and tri-partite interactions with other membrane contacts, including mitochondria and ER at two specific distances, which in my opinion has been more extensivley used to study cell and tissue physiology. They accurately point out that the reversability of this and other systems is challenging and it would be important to define highlight whether the current system allows the study of reversible MERCs. It does not appear as though the reversability of MERCs has been explored in this study. The genetic (PDZD8, FKBP8) and nutritional (starvation) interventions are very helpful to benchmark the system. The description of the methods and data appear to be reproducible and the stastical analyses are acceptable.
Minor comments
As mentioned above, it would be helpful to reference and compare the current study in the context of reversability, which the current MERCdRED system has the potential to provide beyond the state-of-the-art.
The major strength of this study is the development of a stable cell line that allows for the study of MERCs, which has the potential to study the reversible nature of these membrane contact sites. It is debatable as a stable cell line rather than a transient transfection offers a major advancement, even if it does make the study of the system more straightforward, especially if the phenomenon of reversibilty is to be explored. I believe that the CLEM study offers a very informative and precise way to benchmark the ddFP system. Defining how MERC formation and separation (once again the reversibility discovery) have impacts in cell physiology beyond the distances altered by starvation would improve the study. Examining the impact on calcium homeostasis, lipid metabolism, and other aspects of biology that are known to be influenced by MERCs would be interesting. As such, there are no new conceptual, mechanistic, or functional advances, simply minor technical advances in the creation of a stable cell line followed by very solid benchmarking experiments. More complex tri-partite interactions, studied elsewhere, which are conceptually very important for cell and organelle biology, have not been attempted here. Similarly, the notion of studied different types of MERCs, which have been proposed to be important for cell biology, has not been explored using this single reporter. The target audience for this study is one that is interested in membrane contact sites and quantitative biology.
My expertise is in mitochondrial fluorescence imaging and biology. I am not an expert in CLEM.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript entitled "Live-cell imaging reveals nutrient-dependent dynamics of ER-mitochondria contact formation via PDZD8" by Saeko Aoyama-Ishiwatari et al., describes a novel methodology for visualizing contacts between mitochondria and the endoplasmic reticulum (MERCs) by fluorescence microscopy. Inter-organelle contacts, defined as membrane proximities below ~30 nm, fall below the diffraction limit of conventional light microscopy. The method developed by Hirabayashi's laboratory leverages dimerization-dependent fluorescence complementation to create a reporter capable of both visualizing and quantifying ER-mitochondria contacts (MERCs).
Major points:
Minor points:
This timely study provides a valuable and innovative approach to overcoming a longstanding technical limitation in the field, enabling dynamic analysis of ER-mitochondria contacts.
Expertise: cell biology, membrane contact sites, lipid transfer proteins
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
In this study, the authors successfully established a stable cell line expressing MERCdRED, a dimerization-dependent fluorescent protein (ddFP)-based sensor for monitoring mitochondrial-ER contact sites (MERCS). Through light and electron microscopy analyses, they demonstrated that MERCS formation is regulated by nutrient availability and requires PDZD8. While the work is technically sound and well-presented, the biological implications of nutrient-dependent MERCS regulation remain underexplored.
Major Concerns:
Although the manuscript is methodologically robust and suitable for a Methods-type article, its biological significance is limited. The findings primarily serve as proof-of-concept for the MERCdRED tool, without substantially advancing our understanding of MERCS regulation.
To enhance the impact of the study, the authors could use this sensor to investigate novel biological questions-such as the molecular pathways linking nutrient sensing to MERCS dynamics-or explore downstream activities of nutrient-dependent MERCS formation. Deeper mechanistic insights would significantly strengthen the work's contribution to the field.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer #1
Major points
*
In accordance with the reviewers' suggestions, we have incorporated a summary of prior research regarding the developmental origins of dermal fibroblasts into lines 53–56 of the Introduction.
We appreciate your valuable comments regarding the accurate interpretation of TEWL measurements. Estimated TEWL values for human skin have been reported in a systematic review and meta-analysis by Kottner et al. Specifically, the estimated TEWL (95% CI) for individuals aged 18–64 years varies by anatomical site: 15.4 (13.9–17.0) g/m²h for the right cheek, 6.5 (6.2–6.8) g/m²h for the midvolar right forearm, and 36.3 (29.5–43.1) g/m²h for the right palm. In comparison, the TEWL of our EDV model was 9.68 g/m²h, a value relatively close to that of human skin.
We also considered measuring TEWL in artificial skin models lacking epidermis. However, we found that such models remain moist due to culture medium, and pressing the measurement probe against them risks water droplets adhering to the sensor and causing damage. Although we recognize the significance of this measurement as a negative control, we refrained from conducting it due to the limitations of the equipment.
This information has been added to the Results section, lines 178–182.
*
The mechanical measurements in Fig 2 are a nice idea, but it is a bit difficult to interpret without comparison to other conditions (e.g. human skin) or by reporting more universal mechanical parameters (e.g. Young's modulus).*
We greatly appreciate your insightful comments regarding the interpretation of skin viscoelasticity measurements using the Cutometer. The Cutometer is a device that applies negative pressure to the skin to elevate its surface, allowing for the calculation of biomechanical properties based on the temporal changes in skin displacement. Notably, the R7 parameter—defined as the ratio of immediate retraction after pressure release to the maximum deformation during suction—has been shown to correlate significantly with age.
In this study, we evaluated HSEs under the same measurement conditions as those used in previous human clinical studies. Accordingly, we have cited past Cutometer data for human skin and discussed the relationship between those findings and our HSE measurements. These revisions have been made to lines 205–215.
We determined that performing Cutometer measurements on human skin would be impractical due to the ethical committee procedures and associated costs. Although evaluating Young’s modulus using techniques such as AFM to assess the mechanical properties of collagen fibers is a fascinating and informative approach, we have opted not to pursue this analysis due to the substantial time and cost required for sample preparation.
Our response:
Thank you for your valuable advice on strengthening the conclusion of our manuscript. We are currently conducting quantitative analysis through manual counting across multiple fields for all mesenchymal cell markers and Vimentin immunostaining data presented in Fig. 3.
*
Likewise, could the authors clarify whether the cells were passaged before seeding into the HSE, and if so, what passage number. Could passaging affect the responses observed? Please add a discussion point about this.*
Our response:
For all cell types, passage 4 or 5 cells were utilized for the reconstitution of human skin equivalents (HSE). Indeed, Philippeos et al. demonstrated that while CD39, CD90, and CD36 are detectable in primary CD31⁻CD45⁻Ecad⁻ dermal cells, the expression of CD39 is lost after a single passage. In contrast, CD90 and CD36 remain detectable for up to four passages. These findings underscore the impact of in vitro culture on the depletion of fibroblast marker expression. Since we employed NHDFs that had undergone four to five passages for HSE reconstruction, it is reasonable to assume that these cells had already lost specific fibroblast subpopulations, including CD39⁺ cells. Consistent with this, our scRNA-seq analysis revealed that most fibroblasts cultured in 2D formed an artificial population comprising cells in the S and G2M phases, along with secretory-reticular fibroblasts. Additionally, immunohistochemical analysis confirmed a near-complete absence of CD39⁺, CD90⁺, FAP⁺, NG2⁺, and αSMA⁺ cells in the dermis of both D and DV models, further indicating that serial passaging significantly reduces the expression of markers associated with papillary fibroblasts, reticular fibroblasts, and pericytes. Interestingly, the introduction of vascular endothelial cells into the HSE appears to facilitate a partial restoration of fibroblast heterogeneity in cells passaged four to five times. However, whether this effect can be replicated in more extensively passaged fibroblasts remains to be verified. It is well established that excessive passaging induces cellular senescence, leading to reduced proliferative and differentiation capacities in mesenchymal stem cells. Therefore, it is conceivable that fibroblasts beyond a certain passage number may fail to recapitulate dermal mesenchymal cell heterogeneity, even in the presence of endothelial cells.
We have added this discussion to the revised manuscript on lines 372-385, 391–397, and 470-471. However, due to the prolonged culture period required, we regret that we are unable to perform the additional validation experiments at this time.
Our response:
We are currently conducting an additional enrichment analysis on fibroblast subpopulations #0, 1, 2, 6, 8, and 11, identified through UMAP analysis integrating HSE and human skin datasets. We believe that this analysis will elucidate the functional characteristics of each in vitro subpopulation and enable us to speculate on the underlying factors contributing to the observed differences from the human skin analysis results.
Our response:
Thank you for your valuable advice regarding quantitative analysis. We are currently measuring the thickness of the entire epidermal layer, the CK5-positive cell layer, and the CK10-positive cell layer based on HE-stained and IHC-stained images.
Our response:
We are grateful for your insightful comments, which are crucial for a more precise understanding of the physiological relevance of the NP culture model. In response, we are currently undertaking additional analyses to investigate the expression patterns of dermal mesenchymal markers under both NP and AA conditions.
Our response:
As pointed out by reviewers, we acknowledge that elucidating the process and underlying mechanisms by which fibroblasts, whose heterogeneity is compromised in 2D culture, re-differentiate into distinct dermal mesenchymal subtypes constitutes a critical additional analysis to strengthen our findings. Accordingly, we are currently conducting trajectory analysis using Monocle3. This includes identifying branch points that regulate the differentiation of dermal mesenchymal clusters shown in Fig. 4b, as well as predicting transcription factors and cell signaling pathways playing pivotal roles at those branch points. Furthermore, we are planning a CellChat analysis between vascular endothelial cells and dermal mesenchymal cells. We anticipate that integrating the results of these two analyses will provide valuable insights into the differentiation processes of dermal mesenchymal cells, particularly the induction of perivascular cell differentiation.
*
*
Reviewer #1
Minor points
*
Our Response:
Upon thorough consideration, we have deleted the statements that may be regarded as exaggerated (line 26-28 and 346-348).
Our Response:
We appreciate your attention to the reference format error. The necessary revisions have been completed.
Reviewer #2 Major comments:
Our Response
Thank you for your valuable advice, which has helped clarify the physiological phenomena modeled by the NP condition. We are planning additional experiments involving histological analysis, including SA-β-gal staining and the detection of p16^INK4a and/or p21.
Our Response
We sincerely appreciate the important comment regarding the rationale behind the ascorbic acid concentration used in the culture medium. As Reviewer 3 rightly pointed out, concentrations around 100–300 μM are commonly employed in general in vitro assays. In our artificial skin model, we opted for a concentration of 500 μM AA in the growth medium based on two considerations: (1) the model contains a high cell density of approximately 4 × 10⁶ cells immediately after reconstruction, which is expected to result in substantial AA consumption, and (2) AA is not sufficiently stable in culture medium. Given the relatively long medium exchange interval of 48–72 hours, we deemed it necessary to maintain a certain AA level throughout this period. While no rigorous dose–response validation has been conducted, we have confirmed that this concentration does not induce toxicity or abnormalities in skin morphogenesis.
As part of the revision, we considered revisiting the basal medium formulation; however, due to the significant time and resource demands, we have decided to forgo further optimization at this stage.
As described on lines 307–311, the NP medium was formulated to evaluate the potential impact of age-related declines in plasma component transport. We apologize for any confusion regarding the relationship between the HSE growth medium and the NP medium. In response to the reviewer’s suggestion, we have added clarifying explanations and cautionary notes regarding the composition and rationale of these two media in both the Results and Methods sections (line 307-311 and 634-636).
Our response:
As pointed out by reviewers, we acknowledge that elucidating the process and underlying mechanisms by which fibroblasts, whose heterogeneity is compromised in 2D culture, re-differentiate into distinct dermal mesenchymal subtypes constitutes a critical additional analysis to strengthen our findings. Accordingly, we are currently conducting trajectory analysis using Monocle3. This includes identifying branch points that regulate the differentiation of dermal mesenchymal clusters shown in Fig. 4b, as well as predicting transcription factors and cell signaling pathways playing pivotal roles at those branch points. Furthermore, we are planning a CellChat analysis between vascular endothelial cells and dermal mesenchymal cells. We anticipate that integrating the results of these two analyses will provide valuable insights into the differentiation processes of dermal mesenchymal cells, particularly the induction of perivascular cell differentiation. We fully recognize that validation using specific inhibitors is crucial to substantiate the mechanisms suggested by the scRNA-seq analysis. However, given that the reconstruction and reanalysis of the artificial skin model requires more than three months, we have decided not to include these experiments in the current revision and instead consider them as important subjects for future investigation.
Minor comments: 1. The role of pericytes is also underexplored; while their presence is confirmed, functional assays or transcriptomic analyses to elucidate their contribution to ECM remodeling or vascular stability are not fully explored. The origin of pericyte-like cells remains uncertain without lineage tracing or barcoding to distinguish whether they derive from fibroblasts, endothelial cells, or culture artifacts. Since they observe induced differentiation of fibroblast-like cells in 3D culture, it would be compelling to reconstruct differentiation trajectories (pseudotime analysis) from progenitor states to papillary/reticular/pericyte-like states from their scRNAseq data.
Our respnse:
This point will be addressed and validated through our response to Major Comment 3 from Reviewer #2.
Our response
We have planned additional experiments to examine two hypotheses regarding the mechanism underlying the improved responsiveness of the EDV model to AA. The first hypothesis posits that the behavior of ascorbic acid uptake in the cells constituting the EDV model differs from that in the ED model. To investigate this, we plan to analyze the expression patterns of transporter genes potentially involved in the uptake and efflux of ascorbic acid, such as SVCT1 (SLC23A1), SVCT2 (SLC23A2), GLUT1 (SLC2A1), GLUT3 (SLC2A3), GLUT4 (SLC2A4), and MRP4, using scRNA-seq data. The second hypothesis suggests that the absence of bFGF signaling and low FBS treatment under NP conditions may affect subpopulations of dermal mesenchymal cells in the HSEs. To test this, we plan to analyze the expression patterns of dermal mesenchymal cell markers by IHC under NP and AA conditions, following the same approach as shown in Fig. 3.
Our response:
As rightly noted by Reviewer 2, immune cells are integral to skin aging and the maintenance of tissue homeostasis, underscoring the necessity of incorporating them into future research models. Nonetheless, the primary aim of the present study is to elucidate the influence of vascular endothelial cells on dermal mesenchymal cell heterogeneity and to establish an in vitro research model specifically addressing this heterogeneity, with particular emphasis on perivascular cells. Accordingly, we would prefer to consider the analysis of immune cells as a subject for future investigation.
In this study, we opted to use HUVECs as vascular endothelial cells due to their relative ease of expansion in culture. Consequently, we acknowledge the potential limitation in fully recapitulating the functions of tissue-specific endothelial cells. To address this concern, we have revised and expanded the Discussion section on lines 352–356.
Reviewer #3 Major comments:
Upon thorough consideration, we have deleted the statements that may be regarded as exaggerated (line 26-28 and 346-348).
Our response
We are deeply grateful for the reviewer’s constructive feedback. As rightly pointed out, cell ablation and mechanistic assays utilizing signaling inhibitors to assess the contribution of individual mesenchymal subsets are indispensable for reinforcing our findings and claims. However, as the reviewer has also indicated, these experiments would require no less than four months to complete. Consequently, we have opted to forgo high-cost additional experiments such as the optimization of HSE construction protocols and inhibitor-based assays. Instead, we are proactively conducting mechanism-oriented analyses using our existing scRNA-seq and histological datasets. Specifically, we are currently implementing an integrated approach combining Monocle3 and CellChat to pinpoint critical branch points in dermal mesenchymal cell differentiation and to elucidate the signaling pathways orchestrating these bifurcations.
Our Response:
We appreciate your guidance regarding appropriate statistical analysis and data presentation. We planned to revise the depiction of error margins in accordance with best practice guidelines.
Reviewer #3 Minor comments: 1. For Figure 4e, it would be helpful if the authors could clarify in the figure legend or Methods whether the heatmap shows log-normalized expression values (as derived from the Seurat object) or z-scored expression across cells or samples. This distinction affects the interpretation of relative versus absolute expression levels of the collagen and elastic fiber-related genes, which are central to the study's conclusions about ECM remodeling.
Our response:
Thank you for pointing out the inconsistency in data representation. We have revised the manuscript to clearly indicate that Fig. 4e presents the Z-score normalized average expression levels.
*
Our response
Thanks for pointing out the typo, we have corrected it.
Reviewer #4
Minor Points:
In accordance with the reviewer’s suggestion, the display area of the human skin image in Fig. 1c has been modified.
KI67 and TEWL readings for human skin as controls for Fig. 2b-c would help gauge how the organoids perform and whether they are abnormal. What is the elasticity index for facial sagging?
Thank you for your valuable advice, which has deepened our understanding of the evaluation results of HSEs. We are currently planning and conducting an additional analysis by including the quantification of Ki67-positive cells in human skin samples. Regarding the assessment of skin barrier and viscoelasticity using TEWL and Cutometer measurements, we have reffered data from previous clinical studies and added an explanation of the functional differences between HSEs and human skin.
*
Our response:
We appreciate the valuable suggestions provided to investigate the mechanisms underlying the altered VC responsiveness observed in the EDV model. We plan to analyze the expression patterns of transporter genes potentially involved in the uptake and efflux of ascorbic acid, such as SVCT1 (SLC23A1), SVCT2 (SLC23A2), GLUT1 (SLC2A1), GLUT3 (SLC2A3), GLUT4 (SLC2A4), and MRP4, using scRNA-seq data.
There seems to be quite a bit of variability between replicant immunostains, in particular, vimentin in Fig. 3. Can the authors discuss this variability and whether any of the HSE organoid combinations reduced this variability?
Our response:
Thank you for your comments regarding the immunostaining. A reanalysis of the data, including newly acquired immunostaining images during the revision process, is planned.
Our response:
Thank you for your valuable advice. We have added the number of replicates to all figure legends.
Our response:
The photographic data for the EV and ED models in Fig. 1b was incorrect and has therefore been corrected. We sincerely apologize for our oversight. As it was actually the E and EV models that appeared transparent, the description in the text remains unchanged.
Our response:
We sincerely appreciate your insightful guidance regarding the accurate presentation of the histological analysis results. Accordingly, we have revised lines 154–156 in the Results section in line with your recommendations.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript by Kimura et al. define how epidermal morphogenesis in human skin equivalents (HSE) differ by combining vascular endothelial cells, epidermal keratinocytes, and dermal fibroblasts using staining and single-cell RNA-sequencing (scRNA-seq). The three cell system (EDV) displayed higher levels of Ki67+ cells, decreased levels of TEWL, and higher elasticity in comparison to the keratinocyte and fibroblast HSE system (ED). The overall structural morphology between the two systems is quite similar, though the expression of cytokeratin markers varies. EDV organoids specifically express COL1 and COL4 collagen markers surrounding the blood vessels. VEGF-VEGFR1 signaling between endothelia-fibroblasts seems to be pronounced in the EDV organoids according to scRNA-seq, suggesting active signaling between these two cell types. And ascorbic acid appeared to help nutrient poor ED and EDV organoids proliferate compared to controls. This work is well detailed and interesting, helping to define how endothelial cells function to make HSE organoids more faithfully mimic in vivo human skin. Only minor clarifications detailed below are needed.
This work is well detailed and interesting, helping to define how endothelial cells function to make HSE organoids more faithfully mimic in vivo human skin. Only minor clarifications detailed below are needed.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The study develops a tricellular human skin equivalent (HSE) model incorporating epidermal keratinocytes (NHEKs), dermal fibroblasts (NHDFs), and vascular endothelial cells (HUVECs). This model autonomously organizes pericytes, papillary fibroblasts, and reticular fibroblasts, mimicking in vivo dermal mesenchymal heterogeneity. The EDV model (all three cell types) demonstrates enhanced epidermal barrier function (reduced TEWL), dermal elasticity, collagen deposition, and vascular organization compared to simpler models. Single-cell RNA-seq confirms the emergence of pericyte-like and fibroblast subpopulations resembling in vivo counterparts. Nutrient-poor (NP) culture replicates aging phenotypes (reduced proliferation, barrier dysfunction, disordered collagen), rescued by ascorbic acid (AA), highlighting vascular cells' role in skin homeostasis. However, several key methodological clarifications (e.g., heatmap normalization, statistical reporting), more precise qualification of certain claims, and enhanced contextualization within the literature are needed before the work can be considered suitable for publication; I therefore recommend major revision.
Major comments:
Minor comments:
The study innovatively reconstructs dermal mesenchymal heterogeneity using commercially available cells and autonomous tricellular interactions, bypassing costly cell-sorting approaches. This democratizes complex HSE models for broader labs. This study demonstrates that vascularization is critical not only for nutrient supply but for instructing fibroblast/pericyte differentiation and ECM organization. The NP+AA paradigm (Fig. 6) offers a facile in vitro model for skin aging interventions, highlighting AA's efficacy via perivascular mechanisms.
Audience: Tissue engineers, dermatologists, cosmetic/pharma researchers (anti-aging screening), and developmental biologists studying mesenchymal niche regulation.
Placement in existing literature: Recent advances in skin tissue engineering have highlighted the importance of dermal fibroblast heterogeneity in skin homeostasis and regeneration. Single-cell transcriptomic studies (Tabib et al., J Invest Dermatol 2018; Solé-Boldo et al., Commun Biol 2020) have established that papillary and reticular fibroblasts exhibit distinct gene expression and functional roles. Prior engineered skin models incorporating fibroblast subtypes (Moreira et al., Biomater Sci 2023) or pericytes (Paquet-Fifield et al., J Clin Invest 2009) demonstrated improvements in vascularization or epidermal differentiation. However, a unified 3D human skin equivalent integrating vascular cells, pericytes, and spatially organized fibroblast subpopulations has not been systematically achieved. The present work by Kimura et al. advances the field by demonstrating that autonomous interaction among keratinocytes, endothelial cells, pericytes, and heterogeneous fibroblasts significantly enhances both barrier function and dermal elasticity, thus bringing engineered skin models closer to physiological skin. This addresses a key gap between prior single-cell descriptive studies and functional tissue engineering.
Define your field of expertise with a few keywords: experimental dermatology, skin cancer, tissue engineering and 3D skin models, cell biology, tumor microenvironment, and the skin microbiome and barrier function.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this study, the authors present a novel and well-executed approach to reconstructing human skin equivalents (HSEs) that more faithfully replicate the functional complexity of native skin by incorporating the natural heterogeneity of dermal mesenchymal cells, including spatially organized pericytes, papillary fibroblasts, and reticular fibroblasts. Through autonomous interactions among keratinocytes, fibroblasts, and vascular endothelial cells, the fully tricellular EDV model emerged as the most functionally complete among seven engineered HSE variants, demonstrating enhanced epithelialization, barrier integrity, dermal elasticity, and angiogenic architecture. The study's strengths lie in its realistic aging induction via nutrient deprivation by mimicking aspects of vascular insufficiency in the papillary dermis, and its integration of diverse and rigorous evaluation methods, including histological and molecular analyses (Ki67, ECM markers), barrier function (TEWL), and mechanical testing. Notably, ascorbic acid treatment improved epidermal turnover and extracellular matrix organization, particularly through effects on perivascular niche cells, highlighting its translational relevance for anti-aging interventions. Although the EDV model showed superior elasticity via suction testing, more comprehensive mechanical characterization and longitudinal ECM analysis could further elucidate how mesenchymal heterogeneity supports biomechanical resilience. Overall, this work underscores the importance of multicellular crosstalk in skin physiology and positions the EDV model as a robust in vitro platform with high relevance for regenerative medicine, aging research, and therapeutic screening, offering the potential to eliminate animal models in skin biology.
Major comments:
Despite its strengths, the study has several limitations that warrant further investigation. The authors describe a "senescent-like" phenotype under nutrient-poor (NP) conditions, yet do not provide direct evidence of cellular senescence using canonical markers such as SA-β-gal staining, p16^INK4a or p21 expression, or SASP profiling-weakening their aging-related conclusions.
The 500 μM dose of ascorbic acid (AA), while within the reported range for skin models, is at the higher end compared to commonly used concentrations (100-300 μM) and lacks justification via dose/response data. Normal physiological levels and changes in aging dermis should be referenced in discussion. AA is also an additive in their standard HSE media, but this was not sufficiently emphasized to draw attention. Would its removal from the baseline media make a difference? Mechanistically, fibroblast heterogeneity is attributed to keratinocyte and vascular signals, but the signaling pathways involved (e.g., Wnt, TGF-β, VEGF) are not directly examined. Validating which paracrine factors (VEGF, PDGF, LAMA5, KGF) are mediating fibroblast transitions using inhibitors or RNA profiling could shed more light.
Minor comments:
The role of pericytes is also underexplored; while their presence is confirmed, functional assays or transcriptomic analyses to elucidate their contribution to ECM remodeling or vascular stability are not fully explored. The origin of pericyte-like cells remains uncertain without lineage tracing or barcoding to distinguish whether they derive from fibroblasts, endothelial cells, or culture artifacts. Since they observe induced differentiation of fibroblast-like cells in 3D culture, it would be compelling to reconstruct differentiation trajectories (pseudotime analysis) from progenitor states to papillary/reticular/pericyte-like states from their scRNAseq data. Although AA enhanced collagen production and elasticity in the vascularized EDV model, the lack of response in the ED model is not addressed mechanistically. The omission of immune cells which are key players in skin aging and homeostasis could increase physiological relevance of the model. The exclusive use of standard HUVECs may not fully capture the behavior of tissue-specific microvascular endothelial cells, potentially limiting the fidelity of the vascular niche.
This study presents a robust and innovative approach to human skin equivalent (HSE) reconstruction by integrating pericyte-like and endothelial cells with dermal fibroblast subtypes, using only commercially available cell types. A key strength lies in its ability to recapitulate aspects of in vivo fibroblast heterogeneity, including papillary, reticular, and perivascular populations, and to demonstrate functional consequences on tissue architecture, barrier integrity, ECM dynamics, and mechanical properties under aging-like, nutrient-poor conditions. The spontaneous emergence of a pericyte-like population without relying on freshly isolated primary pericytes or complex sorting protocols represents a methodological advance that increases the model's accessibility and scalability. Furthermore, the use of ascorbic acid to reverse aging-associated features in a vascular cell-dependent manner adds a compelling functional dimension, linking cell composition with therapeutic response.
Compared to existing models that either lack vascular cell compartments or do not account for dermal fibroblast heterogeneity, this study fills an important gap at the intersection of skin aging, vascular biology, and mesenchymal-epithelial interactions. The advance is both conceptual by elucidating the role of vascular and perivascular cells in shaping fibroblast identity and function and methodological, through the generation of a human skin model that approximates in vivo complexity without requiring animal models or ethically limited human tissue. The work will be of strong interest to basic science researchers in dermatology, tissue engineering, and aging, and has potential influence in regenerative medicine, cosmetic science, and drug screening, especially in the context of skin repair and anti-aging therapies. The audience is broad but most relevant to specialized communities in skin biology, mesenchymal cell biology, vascular biology, and organoid modeling, and may also attract attention from those developing non-animal testing platforms in applied and translational settings.
As a reviewer with expertise in inflammatory skin disease modeling using both animal systems and 3D organoid cultures, I bring a critical understanding of how cellular composition, microenvironmental cues, and co-culture conditions influence skin physiology and pathology. My interest in developing advanced co-culture systems to recapitulate human skin complexity positions me well to evaluate the relevance, innovation, and translational potential of this vascularized HSE model. I am especially qualified to assess the biological fidelity of the reconstructed skin architecture, the functional outcomes of introducing pericyte-like populations, and the implications of nutrient deprivation and ascorbic acid supplementation as aging-relevant perturbations.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript by Kimura et al investigates the role of different cell populations in the development of human skin equivalents (HSEs). The observe that the addition of vascular endothelial cells to HSEs improves epidermal differentiation and barrier function, alongside differentiation of fibroblasts into papillary, reticular, and pericyte like mesenchymal cells. The authors also use single-cell transcriptomics to characterise the gene signatures and putative signalling pathway in the fibroblasts. Finally, the authors use nutrient poor medium and ascorbic acid to modulate HSE develop.
One of the most significant questions arising from the findings is how the presence of vasculature can induce differentiation of fibroblasts from a common population, especially given that previous studies have shown that fibroblast identity is programmed during development. Some specific comments and suggestions for improving the manuscript are listed below.
Major points:
Minor points:
Overall, the study represents a systematic analysis of how vasculature contributes to skin model development, and the impact on fibroblast differentiation is an interesting observation. It would have been more impactful if some of the pathways and genes were followed up with mechanistic studies, but the findings are still useful to the field. Likewise, further insight into exactly how the vasculature regulates fibroblast phenotype would add to the impact as this is an unexpected but important finding.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer #1 (Evidence, reproducibility and clarity (Required)):*
As stated by the authors in the introduction, the RNA-binding protein Sxl is foundational to understanding sex determination in Drosophila. Sxl has been extensively studied as the master regulator of female sex determination in the soma, where it is known to initiate an alternative splicing cascade leading to the expression of DsxF. Additionally, Sxl has been shown to be responsible for keeping X chromosome dosage compensation off in females, while males hyperactivate their X chromosome. While these roles have been well defined, the authors explore an aspect of Sxl that is quite separate from its role as master regulator of female fate. They describe Sxl-RAC, a Sxl isoform that is expressed in the male and female nervous system. Using several genomic techniques, the authors conclude that the Sxl-RAC isoform associates with chromatin in a similar pattern to the RNA polymerase II/III subunit, Polr3E, and Sxl depends on Polr3E for chromatin-association. Further, neuronal loss of Sxl causes changes in lifetime and geotaxis in a similar manner as loss of Polr3E. The work is thorough and significant and should be appropriate for publication if a few issues can be addressed.
Major Concerns:*
* 1) How physiological is the Sxl chromatin-association assay? As binding interactions are concentration-dependent, how similar is Sxl-DAM expression to wt Sxl expression in neurons? In addition, does the Sxl-DAM protein function as a wt Sxl protein? Does UAS-Sxl-DAM rescue any Sxl loss phenotypes?*
Author response:
As Reviewer 3 correctly notes, Targeted DamID relies on ribosomal re-initiation (codon slippage) to produce only trace amounts of the Dam-fusion protein. By design, this results in expression levels that are significantly lower than those of the endogenous protein. As such, the experiment can be interpreted within a near–wild-type context, rather than as an overexpression model. The primary aim of this experiment was to determine whether Sxl associates with chromatin, and our dataset provides clear evidence supporting such binding.
2) Is Polr3E chromatin-association also dependent on Sxl? They should do the reciprocal experiment to their examination of Sxl chromatin-association in Polr3E knockdown. This might also help address point 1-if wt Sxl is normally required for aspects of Polr3E chromatin binding, then concerns about whether the Sxl-DAM chromatin-association is real or artifactual would be assuaged.
Author response:
This is an interesting thought, however, if Sxl were required for Polr3E recruitment to RNA Pol III, then, in most male Drosophila melanogaster cells, Polr3E would not be incorporated, and males would not be viable (as it is essential for Pol III activity). While it is possible that there could be a subtle effect on Polr3E recruitment, such an experiment, would not alter the central conclusion of our study - that Sxl is recruited to chromatin (accessory to the Pol III complex) via Polr3E.
Minor concerns:
* The observed Sxl loss of function phenotypes are somewhat subtle (although perhaps any behavior phenotype at all is a plus). Did they try any other behaviour assays-courtship, learning/memory, anything else at all to test nervous system function?*
Author response:
Given the exploratory nature of this study, we focused on broader behavioural and transcriptional assays.
While well written, it is sometimes difficult to understand how the experiment was performed or what genotypes were used without looking into the methods sections. One example is they should describe the nature of the Sxl-DAM fusion protein clearly in the results.
Author response:
We will revise these sections to improve clarity and ensure there is no confusion.
* Reviewer #1 (Significance (Required)):
This manuscript represents a dramatic change in our thinking about the action of the Sex-lethal protein. Previously, Sxl was known as the master regulator of both sex determination and dosage compensation, and performed these roles as an RNA-binding protein affecting RNA splicing and translational regulation. Here, the authors describe a sex-non-specific role of Sxl in the male and female nervous system. Further, this activity appears independent of Sxl's RNA binding activity and instead Sxl functions as a chromatin-associating protein working with the RNA pol2/3 factor Polr3E to regulate gene expression. Thus, this represents a highly significant finding. *
Reviewer #2 (Evidence, reproducibility and clarity (Required)):*
Summary: In this paper, the authors report on an unexpected activity for Sex lethal (Sxl) (a known splicing regulator that functions in sex determination and dosage compensation) in binding to chromatin. They show, using DamID, that Sxl binds to approximately the same chromatin regions as Polr3E (a subunit of RNA Pol III). They show that this binding to chromatin is unaffected by mutations in the RNA binding domains or by deletions of either N or C terminal regions of the Sxl protein. This leads the authors to conclude that Sxl must bind to chromatin through some interacting protein working through the central region of the Sxl protein. They show that Sxl binding is dependent on Polr3E function. They show that male-specific neuronal knockdown of Sxl gives similar phenotypes to knockdown of Polr3E in terms of lethality and improved negative geotaxis. They show gene expression changes with knockdown of Sxl in male adult neurons - mainly that metabolic and pigmentation genes go down in expression. They also show that expression of a previously discovered male adult specific form of Sxl (that does not have splicing activity) in the same neurons also leads to changes in gene expression, including more upregulated than downregulated tRNAs. But they don't see (or don't show) that the same tRNA genes are down with knockdown of Sxl. Nonetheless, based on these findings, they suggest that Sxl plays an important role in regulating Pol III activity through the Polr3E subunit.
Major comments:
*
*To be honest, I'm not convinced that the conclusions drawn from this study are correct. The fact that every mutant form of Sxl shows the same result from the DamID labelling is a little concerning. I would like to see independent evidence of the SxlRac protein binding chromatin. *
Do antibodies against this form (or any form) of Sxl bind chromatin in salivary gland polytene chromosomes, for example? Does Sxl from other insects where Sxl has no role in sex determination bind chromatin?
__Author Response: __
Regarding the reviewer’s overall concerns about the legitimacy of the Sxl binding data:
ii) We observed that Sxl binding was significantly reduced upon knockdown of Polr3E, confirming that the signal we observe is biologically specific and not due to technical noise or background. iii) If the concern relates to potential Sxl binding in non-neuronal tissues such as salivary glands, we would like to clarify that all DamID constructs were expressed under elav-GAL4, a pan-neuronal driver. Furthermore, dissections were performed to isolate larval brains, with salivary glands carefully removed. This ensures that chromatin profiles were derived from neuronal tissue exclusively.
iv) Salivary gland polytene chromosome staining with a Sxl antibody in a closely related species (Drosophila virilis) show __binding of Sxl to chromatin __in both sexes (Bopp et al., 1996). We will include more text in the revised manuscript to emphasise these points.
Do antibodies against this form (or any form) of Sxl bind chromatin in salivary gland polytene chromosomes, for example? Does Sxl from other insects where Sxl has no role in sex determination bind chromatin?
Author Response:
Prior work in Drosophila virilis (where Sxl is also required for sex determination and Sxl-RAC is conserved) has already demonstrated Sxl-chromatin association (using a full-length Sxl antibody) in salivary glands using polytene chromosome spreads (Bopp et al., 1996). Binding is observed in both sexes and across the genome, reflecting our observations. We will incorporate this into the revised discussion to support the chromatin-binding role of Sxl across species.
There is a clear and long-overlooked precedent for Sxl's alternative, sex-independent roles, findings that have been largely overshadowed by the gene’s canonical function. Our study not only validates and extends these observations but also brings much-needed attention to this understudied aspect of fundamental biology.
Bopp D, Calhoun G, Horabin JI, Samuels M, Schedl P. Sex-specific control of Sex-lethal is a conserved mechanism for sex determination in the genus Drosophila. Development. 1996 Mar;122(3):971-82. doi: 10.1242/dev.122.3.971. PMID: 8631274.
I would like to see independent evidence of the SxlRac protein binding chromatin.
* *__Author Response: __
We do not believe this is necessary:
iv) Review 3 also believes that this is not necessary (see cross-review below) and highlights the robustness of the Y2H experiments performed by Dong et al., 1999.
Also, given that their DamID experiments reveal that Sxl binds half of the genes encoded in the Drosophila genome, finding that it binds around half of the tRNA genes is perhaps not surprising.
__Author Response: __
Our data show that Sxl binds to a range of Pol III-transcribed loci, and this binding pattern supports the proposed model that Sxl plays a broader regulatory role in Pol III activity. Within these Pol III targets, tRNA genes represent a specific and biologically relevant subset. The emphasis on tRNAs is not to suggest they are the exclusive or primary targets of Sxl, but rather to__ highlight a functionally important class of Pol III-transcribed elements__ that align with the model we are proposing. We will revise the text to better reflect this framing and avoid any confusion regarding the scope of Sxl’s binding profile.
*I would like to see evidence beyond citing a 1999 yeast two-hybrid study that Sxl and Polr3E directly interact with one another. *
Author response:
We do not believe this is necessary (these points were also mentioned above):
In my opinion, the differences in lethality observed with loss of Sxl versus control are unlikely to be meaningful given the different genetic backgrounds. The similar defects in negative geotaxis could be meaningful, but I'm unsure how often this phenotype is observed. What other class of genes affect negative geotaxis? It's a little unclear why having reduced expression of metabolic and pigment genes or of tRNAs would improve neuronal function.
Author response:
While the differences in survival were indeed subtle, they were statistically significant and thus warranted inclusion. Our primary aim in this section was to demonstrate that knockdown of Sxl or Polr3E results in comparable behavioural and transcriptional phenotypes, suggesting overlapping functional roles. In this context, we believe the data were presented transparently and effectively support our interpretation.
Regarding the negative geotaxis phenotype, we appreciate the reviewer’s interest and agree that it is both intriguing and atypical. For this reason, we performed the assay multiple times, particularly in Polr3e knockdowns, to confirm the robustness of the result. To address potential confounding variables, we carefully selected control lines that account for genetic background and transgene insertion site, including KK controls and attP40-matched lines. We also employed multiple independent RNAi lines targeting Sxl to validate the phenotype across different genetic backgrounds.
Although the observed improvement in climbing is unexpected, it is not without precedent in the RNA polymerase III field. Notably, Malik et al. (2024) demonstrated that heterozygous Polr3DEY/+ mutants exhibit a significantly delayed decline in climbing ability with age. We allude to this in the discussion and will revise the text to emphasise this connection more explicitly.
Finally, while we recognise that negative geotaxis is a relatively broad assay and thus does not pinpoint the precise cellular mechanisms involved, we interpret the phenotype as suggesting a neural basis and a functional role for Sxl in the nervous system.
One would expect that not just the same classes of genes would be affected by loss and overexpression of Sxl, but the same genes would be affected - are the same genes changing in opposite directions in the two experiments or just the same classes of genes. Likewise, are the same genes changing expression in the same direction with both Sxl and the Polr3E loss? Also, why are tRNA genes not also affected with Sxl loss. Finally, they describe the changes in gene expression as being in male adult neurons, but the sequencing was done of entire heads - so no way of knowing which cell type is showing differential gene expression.
Author response:
While we do examine gene classes, our approach also includes pairwise correlation analyses of gene expression changes between specific genotypes. Notably, we observed a significant positive correlation between Polr3e knockdowns and Sxl knockdowns, and a significant negative correlation between Sxl-RAC–expressing flies and Sxl knockdowns. Furthermore, we examined Sxl-DamID target genes within our RNA-seq datasets and found a consistent relationship between Sxl targets and genes differentially expressed in Polr3e knockdowns.
Regarding the Pol III qPCR results, we note that tRNA expression changes may require a longer duration of RNAi induction (e.g., beyond 4 days) to become apparent, especially given that phenotypic effects such as changes in lifespan and negative geotaxis only emerge after 20 days or more. It is also plausible that Sxl knockdown leads to a partial reduction in Pol III efficiency, which may not be readily detectable through bulk Pol III qPCRs. We are willing to repeat Pol III qPCRs at later timepoints to further investigate this trend.
Finally, we infer that gene expression changes observed in our RNA-seq data are of neuronal origin, as all knockdown and overexpression constructs used in this study were driven pan-neuronally using elav-/nSyb-GAL4. While we acknowledge that bulk RNA-seq does not provide cell-type resolution, tissue-specific assumptions are widely used in the field when driven by a relevant promoter.
I'm also not sure what I'm supposed to be seeing in panel 5F (or in the related supplemental figure) and if it has any meaning - If they are using the Sxl-T2A-Gal4 to drive mCherry, I think one would expect to see expression since Sxl transcripts are made in both males and in females. Also, one would expect to see active protein expression (OPP staining) in most cells of the adult male brain and I think that is what is observed, but again, I'm not sure what I'm supposed to be looking at given the absence of any arrows or brackets in the figures.
Author Response:
Due to the presence of the T2A tag and the premature stop codon in exon 3 of early male Sxl transcripts, GAL4 expression is not expected in males unless the head-specific SxlRAC isoform is produced. The aim of panel 5F is to demonstrate the spatial overlap between SxlRAC expression (as we are examining male brains) and regions of elevated protein synthesis, as detected by OPP staining.
To quantitatively assess this relationship, we performed colocalisation analysis using ImageJ, which showed a positive correlation between Sxl and OPP signal intensity, supporting this interpretation. It is also evident from our images that regions with lower levels of protein synthesis (such as the neuropil - as shown in independent studies Villalobos-Cantor et al., 2023) concurrently lack Sxl-related signal. We have highlighted regions in Fig. 5 exhibiting higher/lower levels of Sxl/OPP signal to better illustrate this relationship. We can also test the effects of knockdown/overexpression on general protein synthesis if required.
Villalobos-Cantor S, Barrett RM, Condon AF, Arreola-Bustos A, Rodriguez KM, Cohen MS, Martin I. Rapid cell type-specific nascent proteome labeling in Drosophila. Elife. 2023 Apr 24;12:e83545. doi: 10.7554/eLife.83545. PMID: 37092974; PMCID: PMC10125018.
Minor comments:
* Line 223 - 225 - I believe that it is expected that Sxl transcripts would be broadly expressed in the male and female adult, given that it is only the spliced form of the transcript that is female specific in expression. *
As explained above, the only isoform that will be ‘trapped’ by the T2A-GAL4 in males is the Sxl-RAC isoform (as the other isoforms contain premature stop codons). Our immunohistochemistry data indicate that Sxl-RAC is expressed in the male brain, specifically in neurons. Therefore, knockdown experiments in males will reduce all mRNA isoforms, of which, Sxl-RAC is the only one producing a protein.
Line 236 - 238 - Sentence doesn't make sense.
We have addressed and clarified this.
Reviewer #2 (Significance (Required)):
It would be significant to discover that a gene previously thought to function in only sex determination and dosage compensation also moonlights as a regulator of RNA polymerase III activity. Unfortunately, I am not convinced by the work presented in this study that this is the case.
My expertise is in Drosophila biology, including development, transcription, sex determination, morphogenesis, genomics, transcriptomics, DNA binding
Reviewer #3 (Evidence, reproducibility and clarity (Required)):*
Storer, McClure and colleagues use genome-wide DNA-protein binding assays, transcriptomics, and genetics to work out that Drosophila Sxl, widely known as an RNA-binding protein which functions as a splicing factor to determine sex identity in Drosophila and related species, is also a chromatin factor that can stimulate transcription by Pol III and Pol II of genes involved with metabolism and protein homeostasis, specifically some encoding tRNAs.
The evidence for the tenet of the paper -- that Sxl acts as a chromatin regulator with Polr3E, activating at least some of its targets with either Pol III or Pol II -- is logical and compelling, the paper is well written and the figures well presented. Of course, more experiments could always be wished for and proposed, but I think this manuscript could be published in many journals with just a minor revision not involving additional experiments. I have a few specific comments below, all minor.*
Scientific points: - The approach taken for the evaluation of Sxl DNA-binding activity in Fig2 is not entirely clear. I assume these are crosses of elav-Gal4 x different UAS- lines, then using males or females for UAS-Sxl-Full-Length. But what about the others? Were the experiments done in males only? This is hinted at in the main text but not explicitly indicated in the figure or the methods (at least, that I could easily find). And is this approach extended to all other experiments? Longevity? Climbing assays? Considering the role of Sxl, it may be helpful to be fastidiously systematic with this.
Author Response:
We have revised the wording to ensure greater clarity. Males were used for all survival and behavioural experiments (as only males can be leveraged for knocking down Sxl-RAC without affecting the canonical Sxl-F isoform).
- In the discussion, lines 360-61, the authors say: Indeed, knockdown of Polr3E leads to a loss of Sxl binding to chromatin, suggesting a cooperative mechanism. Maybe I am misunderstanding the authors, but when I read "cooperation" in this context I think of biochemical cooperative binding. This is possible, but I do not think a simple 'requirement' test can suggest specifically that this mechanistic feature of biochemical binding is at play. I would expect, for starters, a reciprocal requirement for binding (which is not tested), and some quantitative features that would be difficult to evaluate in vivo. I do not think cooperative binding needs to be invoked anyway, as the authors do not make any specific point or prediction about it. But if they do think this is going on, I think it would need to be referred to as a speculation.
Author Response:
We appreciate that the original wording may have been unclear and will revise the text to more accurately reflect a functional relationship, rather than implying direct cooperation.
- In lines 428-432, the authors discuss the ancestral role of Sxl and make a comparison with ELAV, in the context of an RNA-binding protein that has molecular functions beyond those of a splicing factor, considering the functions of ELAV in RNA stability and translation, and finishing with "suggesting that similar regulatory mechanisms may be at play". I do not understand this latter sentence. Which mechanisms are these? Are the authors referring to the molecular activities of ELAV and SXL? But what would be the similarity? SXL seems to have a dual capacity to bind RNA and protein interactors, which allows it to work both in chromatin-level regulation as well as post-transcriptionally in splicing; but ELAV seems rather to take advantage of its RNA binding function to make it work in multiple RNA-related contexts, all post-transcriptional. I do not see an obvious parallel beyond the fact that RNA binding proteins can function at different levels of gene expression regulation -- but I would not say this parallel are "similar regulatory mechanisms", so I find the whole comparison a bit confusing.
Author Response:
We have reduced this section, as it is largely speculative and intended to highlight potential, though indirect, links in higher organisms. Our goal was primarily to illustrate the possibility that Sxl may have an ancestral role distinct from its well-characterised function, and to suggest a potential avenue for future research into ELAV2’s involvement in chromatin or Pol III regulation.
- One aspect of the work that I find is missing in the discussion is the possibility that the simultaneous capacity of Sxl for RNA binding and Polr3E binding: are these mutually exclusive? if so, are they competitive or hierarchical? how would they be coordinated anyway?
Author Response:
This is an interesting point, and we have expanded on it further in the Discussion section.
- The only aspect of the paper where I found that one could make an experimental improvement is the claim that Sxl induces the expression of genes that have the overall effect of stimulating protein synthesis. The OPP experiment shows a correlation between the expression of Sxl and the rate of protein synthesis initiation. However, a more powerful experiment would be, rather obviously, to introduce Sxl knock-down in the same experiment, and observe whether in Sxl-expressing neurons the incorporation of OPP is reduced. I put this forth as a minor point because the tenet of the paper would not be affected by the results (though the perception of importance of the newly described function could be reinforced).
Author Response:
This could be a valid experiment and we are prepared to perform it if required.
- In a similar way, it would be interesting to know whether the recruitment of Polr3E and Sxl to chromatin is co-dependent or Sxl follows Polr3E. This is also a minor point because this would possibly refine the mechanism of recruitment but does not alter the main discovery.
Author Response:
We have addressed a similar point for Reviewer 2 (see below) and will include a Discussion point for this:
If Sxl were required for Polr3E recruitment to RNA Pol III, then, in most male Drosophila melanogaster cells, Polr3E would not be incorporated, and males would not be viable (as it is essential for Pol III activity). While it is possible that there could be a subtle effect on Polr3E recruitment, such an experiment, would not alter the central conclusion of our study - that Sxl is recruited to chromatin (accessory to the Pol III complex) via Polr3E.
* Figures and reporting:
In Figure 2, it would be helpful to see the truncation coordinate for the N and C truncations.
In Figure 3D, genomic coordinates are missing.
In Figure 3E, the magnitude in the Y axis is not entirely clear (at least not to me). How is the amount of binding across the genome quantified? Is this the average amplitude of normalised TaDa signal across the genome? Or only within binding intervals?
Figure S3E-F: it would be interesting to show the degree of overlap between the downregulated genes that are also binding targets (regardless of the outcome).
Figure 5C-E: similarly to Figure S3, it would be interesting to know how the transcriptional effects compare with the binding targets.
Authors use Gehan-Breslow-Wilcoxon to test survival, which is a bit unusual, as it gives more weight to the early deaths (which are rare in most Drosophila longevity experiments). Is there any rationale behind this? It may be even favour their null hypothesis.*
Author response:
Thank you for the detailed feedback on our figures. We have__ incorporated__ the suggested changes.
We agree that examining the overlap between Sxl binding sites and transcriptional changes is valuable, and we aimed to highlight this in the pie charts shown in Figures S3 and S5. If the reviewer is suggesting a more explicit quantification of the proportion of Sxl-Dam targets with significant transcriptomic changes, we are happy to include this analysis in the final version of the manuscript.
As noted in the Methods, both Gehan–Breslow–Wilcoxon (GBW) and Kaplan–Meier tests were used. The significance in Figure 4a is specific to the GBW test, which we indicated by describing the effect as mild. Our focus here is not on the magnitude of survival differences, but on the consistent trends observed in both Polr3e and Sxl knockdowns.
Writing and language:*
Introduction finishes without providing an outline of the findings (which is fine by me if that is what the authors wanted).
In lines 361-5, the authors say "We speculate that this interaction not only facilitates Pol III transcription but may also influence chromatin architecture and RNA Pol II-driven transcription as observed with Pol III regulation in other organisms". "This interaction" refers to Polr3E-Sxl-DNA interaction and with "Pol III transcription" I presume the authors refer to transcription executed by Pol III. I am not clear about the meaning of the end of the sentence "as observed with Pol III regulation in other organisms". What is the observation, exactly? That Pol III modifies chromatin in Pol II regulated loci, or that Pol III interactors change chromatin architecture?
DPE abbreviation is not introduced (and only used once).
A few typos: Line 41 ...splicing of the Sxl[late] transcripts, which is [ARE?] constitutively transcribed (Keyes et al.,... Line 76 ...sexes but appears restricted to the nervous system [OF] male pupae and adults (Cline et Line 289 ...and S41). To assess any effect [ON]translational output, O-propargyl-puromycin (OPP)o Line 323 ...illustrating that the majority (72%) changes in tRNA levels [ARE] due to upregulation...hi Line 402 ...it was discovered [WE DISCOVERED] Line 792 ...Sxl across chromosomes X, 2 L/R, 3 L/R and 4. The y-axis represents the log[SYMBOL] ratio... This happens in other figure legends as well.*
Author response:
Thank you for the detailed feedback, we have clarified and incorporated the suggested changes.
**Referee Cross-commenting***
Reviewer 1 asks how physiological is the Sxl chromatin-association assay. I think the loss of association in Polr3E knock-down and the lack of association of other splicing factors goes a long way into answering this question. It is true that having positive binding data specifically for Sxl-RAC and negative binding data for a deletion mutant of the RMM domain would provide more robust conclusions (see below), but I am not sure it is completely necessary -- though this will depend on which journal the authors want to send the paper to.
I think that the comment of reviewer 1 about the levels of expression of Sxl-DAM does not apply here because of the way TaDa works - it relies on codon slippage to produce minimal amounts of the DAM fusion protein, so by construction it will be expressed at much lower levels than the endogenous protein.
Reviewer 1 also asks whether Polr3E chromatin-association is also dependent on Sxl, to round up the model and also as a way to address whether Sxl association to chromatin is real. While I agree with this on the former aim (this would be a nice-to-have), I think I disagree on the latter; there is no need for Polr3E recruitment to depend on Sxl for Sxl association to chromatin to be physiologically relevant. Polr3E is a peripheral component of Pol III and unlikely to depend on a factor of restricted expression like Sxl to interact with chromatin. The recruitment of Sxl could well be entirely 'hierarchical' and subject to Polr3E.
Revewer 2 is concerned with the fact that every mutant form of Sxl shows the same result from the DamID labelling. I have to agree with this to a point. A deletion mutant of RMM domains would address this. Microscopy evidence in salivary glands would be nice, certainly, but the system may not lend itself to this particular interaction, which might be short-lived and/or weak. I do not immediately see the relevance of the chromatin binding capacity of non-Drosophilidae Sxl -- though it might indicate that the impact of the discovery is less likely to go beyond this group.
Reviewer 2 does not find surprising that some tRNA genes (less than half) are regulated by Sxl. I think the value of that observation is just qualitative, as tRNAs are Pol III-produced transcripts, but their point is correct. A hypergeometric test could settle this.
Reviewer 2 is concerned that the evidence of direct interaction between Sxl and Polr3E is a single 1999 two-hybrid study. But that paper contains also GST pull-downs that narrow down the specific domains that mediate binding, and perform the binding in competitive salt conditions. I think it is enough. The author team, I think, are not biochemists, so finding the right collaborators and performing these experiments would take time that I am not sure is warranted.
Reviewer 2 is also concerned that the longevity assays may not be meaningful due to the difference in genetic backgrounds. This is a very reasonable concern (which I would extend to the climbing assays - any quantitative phenotype is sensitive to genetic background). However, I think the authors here may have already designed the experiment with this in mind - the controls express untargeted RNAi constructs, but I lose track of which one is control of which. This should be clarified in Methods.
Other comments are in line, I think, with what I have pointed out and I generally agree with everything else that has been said.
Reviewer #3 (Significance (Required)):
Drosophila Sxl is widely known as an RNA-binding protein which functions as a splicing factor to determine sex identity in Drosophila and related species. It is a favourite example of how splicing factors and alternative can have profound influence in biology and used cleverly in the molecular circuitry of the cell to enact elegant regulatory decisions.
In this work, Storer, McClure and colleagues use genome-wide DNA-protein binding assays, transcriptomics, and genetics to work out that Sxl is also a chromatin factor with an sex-independent, neuron-specific role in stimulating transcription by Pol III and Pol II, of genes involved with metabolism and protein homeostasis, including some encoding tRNAs.
This opens a large number of interesting biological questions that range from biochemistry, gene regulation or neurobiology to evolution. How is the simultaneous capacity of binding RNA and chromatin (with the same protein domain, RRM) regulated/coordinated? How did this dual activity evolve and which one is the ancestral one? How many other RRM-containin RNA-binding proteins can also bind chromatin? How is Sxl recruited to chromatin to both Pol II and Pol III targets and are they functionally related? If so, how is the coordination of cellular functions activated through different RNA polymerases taking place and what is the role of Sxl in this? What are the functional consequences to neuronal biology? Does this affect similarly all Sxl-expressing neurons?
The evidence for the central tenet of the paper -- that Sxl acts as a chromatin regulator with Polr3E, activating at least some of its targets with either Pol III or Pol II -- is logical and compelling, the paper is well written and the figures well presented. Of course, more experiments could always be wished for and proposed, but I think this manuscript could be published in many journals with just a minor revision not involving additional experiments.*
Reviewer #4 (Evidence, reproducibility and clarity (Required)):
*The convincing analysis demonstrates a role for the Drosophila Sex determining gene sex lethal in controlling aspects of transcription in the nervous system independent of its role in splicing. Interaction with an RNA Pol III subunit mediating Sxl association with chromatin and similar knockdown phenotypes strongly support the role of Sxl in the regulation of neuronal metabolism. Given that Sxl is an evolutionary recent acquisition for sex determination, the study may reveal an ancestral role for Sxl.
The conclusions are well justified by the datasets presented and I have no issues with the study or the interpretation. Throughout the work is well referenced, though perhaps the authors might take a look at Zhang et al (2014) (PMID: 24271947) for an interesting evolutionary perspective for the discussion.*
Author Response:
Thank you for the thoughtful suggestion. We will be sure to incorporate the findings from Zhang et al. regarding the evolution of the sex determination pathway.
*I have some minor comments for clarification:
There is no Figure 2b, should be labelled 2 or label TaDa plots as 2b
Clarify if Fig 2 data are larval or adult *
*Larval
Fig 3d - are these replicates or female and male?
Please elaborate on tub-GAL80[ts] developmental defects
Fig 4e, are transcriptomics done with the VDRC RNAi line? The VDRC and BDSC RNAi lines exhibit different behaviours - former has "better" survival and Better negative geotaxis, the latter seems to have poorer survival but little geotaxis effect?*
*Fig S3 - volcano plot for Polr3E?
Fig S4a - legend says downregulated genes?
The discussion should at least touch on the fact that Sxl amorphs (i.e. Sxl[fP7B0] are male viable and fertile, emphasising that the newly uncovered role is not essential.*
Author Response:
We agree with the suggestions outlined in the comments and have made the appropriate revisions.
Reviewer #4 (Significance (Required)):*
A nonessential role for Sxl in the nervous system independent of sex-determination contributes to better understanding a) the evolution of sex determining mechanisms, b) the role of RNA PolIII in neuronal homeostasis and c) more widely to the neuronal aging field. I think this well-focused study reveals a hitherto unsuspected role for Sxl.*
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The convincing analysis demonstrates a role for the Drosophila Sex determining gene sex lethal in controlling aspects of transcription in the nervous system independent of its role in splicing. Interaction with an RNA Pol III subunit mediating Sxl association with chromatin and similar knockdown phenotypes strongly support the role of Sxl in the regulation of neuronal metabolism. Given that Sxl is an evolutionary recent acquisition for sex determination, the study may reveal an ancestral role for Sxl.
The conclusions are well justified by the datasets presented and I have no issues with the study or the interpretation. Throughout the work is well referenced, though perhaps the authors might take a look at Zhang et al (2014) (PMID: 24271947) for an interesting evolutionary perspective for the discussion. I have some minor comments for clarification:
There is no Figure 2b, should be labelled 2 or label TaDa plots as 2b
Clarify if Fig 2 data are larval or adult
Fig 3d - are these replicates or female and male?
Please elaborate on tub-GAL80[ts] developmental defects
Fig 4e, are transcriptomics done with the VDRC RNAi line? The VDRC and BDSC RNAi lines exhibit different behaviours - former has "better" survival and Better negative geotaxis, the latter seems to have poorer survival but little geotaxis effect?
Fig S3 - volcano plot for Polr3E?
Fig S4a - legend says downregulated genes?
The discussion should at least touch on the fact that Sxl amorphs (i.e. Sxl[fP7B0] are male viable and fertile, emphasising that the newly uncovered role is not essential
A nonessential role for Sxl in the nervous system independent of sex-determination contributes to better understanding a) the evolution of sex determining mechanisms, b) the role of RNA PolIII in neuronal homeostasis and c) more widely to the neuronal aging field. I think this well-focused study reveals a hitherto unsuspected role for Sxl.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Storer, McClure and colleagues use genome-wide DNA-protein binding assays, transcriptomics, and genetics to work out that Drosophila Sxl, widely known as an RNA-binding protein which functions as a splicing factor to determine sex identity in Drosophila and related species, is also a chromatin factor that can stimulate transcription by Pol III and Pol II of genes involved with metabolism and protein homeostasis, specifically some encoding tRNAs.
The evidence for the tenet of the paper -- that Sxl acts as a chromatin regulator with Polr3E, activating at least some of its targets with either Pol III or Pol II -- is logical and compelling, the paper is well written and the figures well presented. Of course, more experiments could always be wished for and proposed, but I think this manuscript could be published in many journals with just a minor revision not involving additional experiments. I have a few specific comments below, all minor.
Scientific points:
Figures and reporting:
Writing and language:
Referee Cross-commenting
Reviewer 1 asks how physiological is the Sxl chromatin-association assay. I think the loss of association in Polr3E knock-down and the lack of association of other splicing factors goes a long way into answering this question. It is true that having positive binding data specifically for Sxl-RAC and negative binding data for a deletion mutant of the RMM domain would provide more robust conclusions (see below), but I am not sure it is completely necessary -- though this will depend on which journal the authors want to send the paper to.
I think that the comment of reviewer 1 about the levels of expression of Sxl-DAM does not apply here because of the way TaDa works - it relies on codon slippage to produce minimal amounts of the DAM fusion protein, so by construction it will be expressed at much lower levels than the endogenous protein.
Reviewer 1 also asks whether Polr3E chromatin-association is also dependent on Sxl, to round up the model and also as a way to address whether Sxl association to chromatin is real. While I agree with this on the former aim (this would be a nice-to-have), I think I disagree on the latter; there is no need for Polr3E recruitment to depend on Sxl for Sxl association to chromatin to be physiologically relevant. Polr3E is a peripheral component of Pol III and unlikely to depend on a factor of restricted expression like Sxl to interact with chromatin. The recruitment of Sxl could well be entirely 'hierarchical' and subject to Polr3E.
Revewer 2 is concerned with the fact that every mutant form of Sxl shows the same result from the DamID labelling. I have to agree with this to a point. A deletion mutant of RMM domains would address this. Microscopy evidence in salivary glands would be nice, certainly, but the system may not lend itself to this particular interaction, which might be short-lived and/or weak. I do not immediately see the relevance of the chromatin binding capacity of non-Drosophilidae Sxl -- though it might indicate that the impact of the discovery is less likely to go beyond this group.
Reviewer 2 does not find surprising that some tRNA genes (less than half) are regulated by Sxl. I think the value of that observation is just qualitative, as tRNAs are Pol III-produced transcripts, but their point is correct. A hypergeometric test could settle this.
Reviewer 2 is concerned that the evidence of direct interaction between Sxl and Polr3E is a single 1999 two-hybrid study. But that paper contains also GST pull-downs that narrow down the specific domains that mediate binding, and perform the binding in competitive salt conditions. I think it is enough. The author team, I think, are not biochemists, so finding the right collaborators and performing these experiments would take time that I am not sure is warranted.
Reviewer 2 is also concerned that the longevity assays may not be meaningful due to the difference in genetic backgrounds. This is a very reasonable concern (which I would extend to the climbing assays - any quantitative phenotype is sensitive to genetic background). However I think the authors here may have already designed the experiment with this in mind - the controls expres untargeted RNAi constructs, but I lose track of which one is control of which. This should be clarified in Methods.
Other comments are in line, I think, with what I have pointed out and I generally agree with everything else that has been said.
Drosophila Sxl is widely known as an RNA-binding protein which functions as a splicing factor to determine sex identity in Drosophila and related species. It is a favourite example of how splicing factors and alternative can have profound influence in biology and used cleverly in the molecular circuitry of the cell to enact elegant regulatory decisions.
In this work, Storer, McClure and colleagues use genome-wide DNA-protein binding assays, transcriptomics, and genetics to work out that Sxl is also a chromatin factor with an sex-independent, neuron-specific role in stimulating transcription by Pol III and Pol II, of genes involved with metabolism and protein homeostasis, including some encoding tRNAs.
This opens a large number of interesting biological questions that range from biochemistry, gene regulation or neurobiology to evolution. How is the simultaneous capacity of binding RNA and chromatin (with the same protein domain, RRM) regulated/coordinated? How did this dual activity evolve and which one is the ancestral one? How many other RRM-containin RNA-binding proteins can also bind chromatin? How is Sxl recruited to chromatin to both Pol II and Pol III targets and are they functionally related? If so, how is the coordination of cellular functions activated through different RNA polymerases taking place and what is the role of Sxl in this? What are the functional consequences to neuronal biology? Does this affect similarly all Sxl-expressing neurons?
The evidence for the central tenet of the paper -- that Sxl acts as a chromatin regulator with Polr3E, activating at least some of its targets with either Pol III or Pol II -- is logical and compelling, the paper is well written and the figures well presented. Of course, more experiments could always be wished for and proposed, but I think this manuscript could be published in many journals with just a minor revision not involving additional experiments.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
In this paper, the authors report on an unexpected activity for Sex lethal (Sxl) (a known splicing regulator that functions in sex determination and dosage compensation) in binding to chromatin. They show, using DamID, that Sxl binds to approximately the same chromatin regions as Polr3E (a subunit of RNA Pol III). They show that this binding to chromatin is unaffected by mutations in the RNA binding domains or by deletions of either N or C terminal regions of the Sxl protein. This leads the authors to conclude that Sxl must bind to chromatin through some interacting protein working through the central region of the Sxl protein. They show that Sxl binding is dependent on Polr3E function. They show that male-specific neuronal knockdown of Sxl gives similar phenotypes to knockdown of Polr3E in terms of lethality and improved negative geotaxis. They show gene expression changes with knockdown of Sxl in male adult neurons - mainly that metabolic and pigmentation genes go down in expression. They also show that expression of a previously discovered male adult specific form of Sxl (that does not have splicing activity) in the same neurons also leads to changes in gene expression, including more upregulated than downregulated tRNAs. But they don't see (or don't show) that the same tRNA genes are down with knockdown of Sxl. Nonetheless, based on these findings, they suggest that Sxl plays an important role in regulating Pol III activity through the Polr3E subunit.
Major comments:
To be honest, I'm not convinced that the conclusions drawn from this study are correct. The fact that every mutant form of Sxl shows the same result from the DamID labelling is a little concerning. I would like to see independent evidence of the SxlRac protein binding chromatin. Do antibodies against this form (or any form) of Sxl bind chromatin in salivary gland polytene chromosomes, for example? Does Sxl from other insects where Sxl has no role in sex determination bind chromatin?
Also, given that their DamID experiments reveal that Sxl binds half of the genes encoded in the Drosophila genome, finding that it binds around half of the tRNA genes is perhaps not surprising.
I would like to see evidence beyond citing a 1999 yeast two-hybrid study that Sxl and Polr3E directly interact with one another. In my opinion, the differences in lethality observed with loss of Sxl versus control are unlikely to be meaningful given the different genetic backgrounds. The similar defects in negative geotaxis could be meaningful, but I'm unsure how often this phenotype is observed. What other class of genes affect negative geotaxis? It's a little unclear why having reduced expression of metabolic and pigment genes or of tRNAs would improve neuronal function.
One would expect that not just the same classes of genes would be affected by loss and overexpression of Sxl, but the same genes would be affected - are the same genes changing in opposite directions in the two experiments or just the same classes of genes. Likewise, are the same genes changing expression in the same direction with both Sxl and the Polr3E loss? Also, why are tRNA genes not also affected with Sxl loss. Finally, they describe the changes in gene expression as being in male adult neurons, but the sequencing was done of entire heads - so no way of knowing which cell type is showing differential gene expression.
I'm also not sure what I'm supposed to be seeing in panel 5F (or in the related supplemental figure) and if it has any meaning - If they are using the Sxl-T2A-Gal4 to drive mCherry, I think one would expect to see expression since Sxl transcripts are made in both males and in females. Also, one would expect to see active protein expression (OPP staining) in most cells of the adult male brain and I think that is what is observed, but again, I'm not sure what I'm supposed to be looking at given the absence of any arrows or brackets in the figures.
Minor comments:
Line 223 - 225 - I believe that it is expected that Sxl transcripts would be broadly expressed in the male and female adult, given that it is only the spliced form of the transcript that is female specific in expression.
Line 236 - 238 - Sentence doesn't make sense.
It would be significant to discover that a gene previously thought to function in only sex determination and dosage compensation also moonlights as a regulator of RNA polymerase III activity. Unfortunately, I am not convinced by the work presented in this study that this is the case.
My expertise is in Drosophila biology, including development, transcription, sex determination, morphogenesis, genomics, transcriptomics, DNA binding
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
As stated by the authors in the introduction, the RNA-binding protein Sxl is foundational to understanding sex determination in Drosophila. Sxl has been extensively studied as the master regulator of female sex determination in the soma, where it is known to initiate an alternative splicing cascade leading to the expression of DsxF. Additionally, Sxl has been shown to be responsible for keeping X chromosome dosage compensation off in females, while males hyperactivate their X chromosome. While these roles have been well defined, the authors explore an aspect of Sxl that is quite separate from its role as master regulator of female fate. They describe Sxl-RAC, a Sxl isoform that is expressed in the male and female nervous system. Using several genomic techniques, the authors conclude that the Sxl-RAC isoform associates with chromatin in a similar pattern to the RNA polymerase II/III subunit, Polr3E, and Sxl depends on Polr3E for chromatin-association. Further, neuronal loss of Sxl causes changes in lifetime and geotaxis in a similar manner as loss of Polr3E. The work is thorough and significant and should be appropriate for publication if a few issues can be addressed.
Major Concerns
Minor concerns
The observed Sxl loss of function phenotypes are somewhat subtle (although perhaps any behavior phenotype at all is a plus). Did they try any other behaviour assays-courtship, learning/memory, anything else at all to test nervous system function?
While well written, it is sometimes difficult to understand how the experiment was performed or what genotypes were used without looking into the methods sections. One example is they should describe the nature of the Sxl-DAM fusion protein clearly in the results.
This manuscript represents a dramatic change in our thinking about the action of the Sex-lethal protein. Previously, Sxl was known as the master regulator of both sex determination and dosage compensation, and performed these roles as an RNA-binding protein affecting RNA splicing and translational regulation. Here, the authors describe a sex-non-specific role of Sxl in the male and female nervous system. Further, this activity appears independent of Sxl's RNA binding activity and instead Sxl functions as a chromatin-associating protein working with the RNA pol2/3 factor Polr3E to regulate gene expression. Thus, this represents a highly significant finding.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
*Here we provide a point-by-point reply describing the revisions already carried out and included in the transferred manuscript. *
Reviewer #1 – Evidence, reproducibility and clarity
This is a rigorous biophysical characterization of a protein-protein interaction relevant to CDA-1 disease. The two proteins were purified in an E. coli host but CD and DLS was performed to ensure that the purified protein is well folded. An impressive native protein EMSA was used to show a 1:1 complex. While common for protein-nucleic acid complexes, EMSAs are much more challenging with protein complexes. A higher-running complex, likely a heterotetramer was implied at higher protein concentrations. These results were supported with SEC-MALS analysis and analytic ultracentrifugation analysis. Thermophoresis and ITC were used to report a nanomolar affinity of the proteins for each other. SEC-SAXS supported the conclusions about stoichiometry and composition inferred from the earlier methods and suggested that the dimerization interface comes from CDIN1. Next HDX-MS was used to identify putative interface residues, which were then mutated in each of the proteins and assessed for binding using coimmunoprecipitation. This study uses at least 10 orthogonal biophysical and/or biochemical methodologies to characterize an important protein-protein interaction and the analysis is clear and so is the writing. I couldn't (reading it once) find any grammatical or other errors in the text or figures. This manuscript is top-quality and suitable for publication.
__Reviewer #1 – Significance __
Such detailed structural and mechanistic studies are greatly lacking in many clinical conditions for which mutations are known (unless they cause cancer, neurodegenerative disease, and so on). We need more such studies on disease topics! This study will be of interest to the hematologic diseases community.
1. Response – ____Significance
We thank Reviewer #1 for the thoughtful and encouraging evaluation of our work. We are particularly grateful for recognizing the significance of studying protein-protein interaction in the context of CDA-I disease, as well as the rigor and clarity of our biophysical and biochemical characterization.
We appreciate the reviewer's acknowledgment of the challenges associated with native protein EMSAs. We are pleased that our use of multiple orthogonal techniques was recognized as a strength of the study. We are gratified that the comprehensiveness and coherence of our data and the manuscript's clarity were well received.
We thank the reviewer for noting the broader impact of our findings on the hematologic disease community. As highlighted, there is a pressing need for a mechanistic understanding of non-oncologic, non-neurodegenerative diseases, and our studies address this gap.
We are honored by the reviewer's endorsement of our manuscript as "top-quality and suitable for publication". We value the reviewer's highly supportive and motivating feedback.
__Reviewer #2 – 1. Evidence, reproducibility and clarity __
This manuscript presents structural and biochemical characterization of the interaction between CDIN1 and the C-terminal domain of Codanin1, shedding light on a complex implicated in Congenital Dyserythropoietic Anemia Type I (CDA-I). While the authors provide valuable structural insights and identify disease-associated mutations that impair CDIN1-Codanin1 binding, I think several important concerns should be addressed to strengthen both the mechanistic claims and their functional relevance.
Contradiction Between Stoichiometry Models:
The authors propose that CDIN1 and Codanin1Cterm primarily form a heterodimer in vitro. However, this appears to contradict previous reports indicating a tetra-heteromeric arrangement. Additionally, while CDIN1 homodimerize seems confusing to me, do the authors suggest it is stable without Codanin1? This seems contrary to findings that CDIN1 is unstable in the absence of Codanin1 (Sedor, S.F., Shao, S. nature comm 2025, Swickley, G., Bloch, Y., Malka, L. et al 2020 BMC Mol and Cell Biol). These inconsistencies raise concerns about whether the observed stoichiometries are physiologically relevant or artifacts of in vitro reconstitution, especially since full-length Codanin1 was not studied.
2.1 Response ____– Consistent stoichiometry of Codanin1Cterm
We thank Reviewer #2 for raising critical points regarding the stoichiometry and physiological relevance of the CDIN1-Codanin1 interaction. The following response clarifies the rationale and interpretation in relation to previous findings.
Stoichiometry of CDIN1-Codanin1Cterm complex:
Recent Cryo-EM studies of full-length Codanin1 (Jeong, Frater et al. 2025, Sedor and Shao 2025) suggest independent internal dimerization domains (452-798 and 841-1000 amino acid residue) driving homodimer formation, with each Codanin1 monomer binding one CDIN1 via the C-terminal region (1005-1227 amino acid residue), resulting in a tetra-heteromeric complex. Therefore, the complete assembly appears as a dimer of heterodimers in the full-length context.
In our study, Codanin1 was truncated to retain only the CDIN1-binding C-terminus (1005-1227 amino acid residues), eliminating the homodimerization ability of Codanin1. Hence, in the case of truncated Codanin1Cterm, the minimal complex we observe is a 1:1 heterodimer of CDIN1-Codanin1Cterm, which is fully consistent with the equimolar stoichiometry of CDIN1-Codanin1 complex seen in the full-length structure.
Stability and oligomeric state of CDIN1 in the absence of Codanin1:
We concur with the reviewer that Sedor et al. (2025) and Swickley et al. (2020) reported decreased CDIN1 levels in cells lacking Codanin1, implying in vivo dependence of CDIN1 on Codanin1 partner for stability (Swickley, Bloch et al. 2020, Sedor and Shao 2025). The purified CDIN1 is monodisperse (Supplementary Figure 2D), exhibits thermal stability with a melting temperature of 48 °C (Supplementary Figure 2E), and displays proper folding as indicated by CD measurements (Supplementary Figure 2B). Additionally, SAXS profiles of CDIN1 correspond to AlphaFold predictions (Fig. 2B). Together, our findings indicate that the recombinant CDIN1 forms a stable conformation in vitro without Codanin1. To the best of our knowledge, no previous research has directly identified the endogenous oligomeric states of CDIN1 within cellular content.
We fully acknowledge that future analysis of the full-length Codanin1-CDIN1 assembly in a cellular context will be necessary for understanding physiological stoichiometries. As outlined in the General statements, our study focuses on the C-terminus of Codanin1 to describe the binding interface and complex biophysical properties of the CDIN-Codanin1Cterm complex.
__Reviewer #2 – ____2. Unvalidated Functional Claims: __
The manuscript identifies several CDA-I-associated mutations that disrupt CDIN1-Codanin1 interaction. However, the authors do not test how these mutations affect the biological function of the complex, particularly its role in ASF1 sequestration or histone trafficking. Given the central importance of this axis in their disease model, functional validation (e.g., ASF1 localization, histone deposition assays) is necessary to support these mechanistic conclusions.
2.2 Response – ____Hypothetical model as discussion merit
We thank the reviewer for the comment regarding the functional implications of CDA-I-associated mutations and their potential impact on ASF1 sequestration and histone trafficking hypothesized within the Discussion. We fully agree that understanding the downstream biological consequences of disrupted CDIN1-Codanin1 interaction is critical for elucidating the full molecular basis of CDA-I pathogenesis.
In the Future research directions of the Discussion, we have acknowledged and emphasized the need for follow-up studies using erythroblast cell lines to determine whether specific disease-associated mutations disrupt CDIN1-Codanin1 binding, leading to functional defects relevant to erythropoiesis and nuclear architecture typical for CDA-I disease.
However, as we respectfully note in General Statements, the main aim of the present study was to provide a rigorous biophysical characterization of the CDIN1-Codanin1Cterm interaction. Proposed cellular experiments, though relevant, are beyond the conceptual scope of the presented studies.
Reviewer #2 – ____3. Speculative and Potentially Contradictory Model:
The proposed model suggests that CDIN1 competes with ASF1 for Codanin1 binding, thereby indirectly promoting histone delivery to the nucleus. However, emerging data indicate that Codanin1, CDIN1, and ASF1 can form a stable ternary complex, calling into question this competitive binding hypothesis (Sedor, S.F., Shao, S. nature comm 2025). The authors do not acknowledge or discuss these findings, and the model in its current form may therefore be oversimplified or inaccurate.
2.3 Response – ____Hypothetical model fully aligned with current knowledge
We fully acknowledged and discussed in the current manuscript the recent findings demonstrating that Codanin1, CDIN1, and ASF1 can form a ternary complex (Sedor, S.F., Shao, S. Nature Comm. 2025; Jeong, T. K. et al. Nature Comm. 2025). Our revised model was updated accordingly to reflect the collaborative binding of Codanin1, CDIN1, and ASF1, and is presented in alignment with published data.
While earlier versions of our work published on the BioRxiv server (May 26, 2023) proposed a competitive hypothesis, the current manuscript incorporates recent literature and prior reviewer feedback to offer a refined model. We believe that the updated hypothesis suggests a plausible mechanism for how CDIN1 modulates Codanin1 function, which will be further tested in future cellular studies.
Reviewer #2 – 4. Significance:
Overall, the study adds to our structural understanding of CDIN1 and Codanin1 interactions, but the functional interpretations are currently speculative, and in some cases in conflict with existing literature. The manuscript would benefit significantly from addressing these discrepancies, incorporating relevant data on ASF1, and clarifying whether the observed assemblies reflect physiological complexes.
__2.4 Response – Significance __
We thank Reviewer #2 for the constructive feedback. As noted in General Statements, our current manuscript is primarily dedicated to defining the molecular architecture and interactions of the CDIN1–Codanin1Cterm core interface. We agree that follow-up ASF1‑dependent functional assays will be critical to fully validate observed assemblies, but these experiments lie outside the scope of the present study and are ongoing in our laboratory.
To address the reviewer's concern about possible speculative interpretation, we have:
__Reviewer #3 – Evidence, reproducibility and clarity: __
Congenital Dyserythropoietic Anemia Type I (CDA I) is an autosomal recessive disorder characterized by ineffective erythropoiesis and distinctive nuclear morphology ("Swiss cheese" heterochromatin) in erythroblasts. CDA I is caused by mutations in CDAN1 and CDIN1. Codanin1, encoded by CDAN1, is part of the cytosolic ASF1-H3.1-H4-Importin-4 complex, which regulates histone trafficking to the nucleus. CDIN1 has been shown to bind the C-terminal domain of Codanin-1, but until now, pathogenic mutations had not been directly linked to the disruption of this interaction.
In this study, the authors used biophysical techniques to characterize the interaction between Codanin-1's C-terminal region (residues 1005-1227) and CDIN1, demonstrating high-affinity, equimolar binding. HDX-MS identified interaction hotspots, and disease-associated mutations in these regions disrupted complex formation. The authors propose that such disruption prevents ASF1 sequestration in the cytoplasm, thereby reducing nuclear histone levels and contributing to the chromatin abnormalities seen in CDA I.
Major Comments:
1. Use of Codanin-1 Fragment:
Most experiments were conducted using only the C-terminal 223 amino acids of Codanin-1. While this region is known to bind CDIN1, it is unclear whether its conformation is maintained in the context of the full-length protein. This could affect binding properties and structural interpretations. The authors should discuss how structural differences between the isolated C-terminus and the full-length Codanin-1 may influence the conclusions.
Response of authors ____#3
3.1 Response: Use of Codanin-1 Fragment as biding part to CDIN1
We thank the reviewer for the important observation regarding the use of the C-terminal fragment of Codanin1. As noted in the manuscript (e.g., p. 30, line 721 and p. 32, line 761), we fully acknowledge that the truncation of Codanin1 may influence its conformational dynamics or contextual folding relative to the full-length protein.
However, several lines of evidence suggest that the C-terminal 223 amino acid residues—responsible for CDIN1 binding—are structurally autonomous and have minimal intramolecular contacts with upstream regions. Published cryo-EM and biochemical data (Jeong, Frater et al. 2025, Sedor and Shao 2025), in conjunction with AlphaFold structural predictions (Fig. 2D) and our co-immunoprecipitation assays (Fig. 3F), consistently support a model wherein the CDIN1-binding region is flexible and spatially isolated from the core structural domains of Codanin1. Additionally, results from our co-immunoprecipitation assay (Fig. 3F) indicate that full-length Codanin1 and truncated Codanin1Cterm interact with CDIN1 similarly, further supporting the isolated manner of the C-terminal fragment. The available data together imply that the C-terminal fragment used in our study retains its native conformation and binding properties when expressed independently.
While our findings are confined to the interaction domain and do not reflect full-length Codanin1’s architecture, we believe the use of the C-terminal minimal fragment of Codanin1 enables precise dissection of the CDIN1-binding interface and yields mechanistic insights without introducing significant structural artifacts.
We agree with the reviewer that future work incorporating full-length Codanin1, especially in a cellular context, will be instrumental to fully characterize higher-order assembly and regulatory functions.
__Reviewer #3 – 2. ____Graphical Abstract and Domain Independence: __
The graphical abstract presents the Codanin-1 C-terminus as an independent domain, but no direct evidence is provided to support its structural autonomy in vivo.
The authors should clarify whether the C-terminal region functions as a distinct domain in the context of the full-length protein.
__3.2 Response –____ Independent C-terminal domain __
We thank the reviewer for bringing up the question of the independence of the C-terminal domain. Although direct in vivo proof of C-terminal autonomy is not yet available, published cryo-EM structures of full-length Codanin1, our biophysical characterization, and AlphaFold models all consistently indicate that the C-terminal 223 amino acid residues of Codanin1 form a structurally independent binding module. In the graphical abstract, we illustrated the C‑terminal domain as a loosely connected part of Codanin1 to highlight its independence and to emphasize the specific focus of our studies.
To articulate limitations of our studies focused on the C-terminal part of Codanin1, we stated in the Functional implications of CDA-I-related mutations in the Discussion, p. 30, l. 721-724: “However, our measurements do not exclude the possible role of the disordered regions in full-length Codanin1. For example, CDIN1 could potentially stabilize full-length Codanin1 by rearranging the disordered regions into a more condensed structure, thereby augmenting the structural stability of Codanin1.”
Reviewer #3 – 3.____Pathogenic Mutations Beyond the Binding Site:
The study highlights a triplet mutation that impairs CDIN1 binding. However, most CDA I‑associated mutations in CDAN1 are dispersed across the entire protein and may not affect CDIN1 interaction directly.
The authors should discuss alternative mechanisms by which mutations in other regions of Codanin-1 might cause disease.
3.3 Response – Pathogenic mutations outside the binding site – alternative mechanisms
We appreciate the reviewer noting that most CDA-I-associated CDAN1 mutations are outside the CDIN1-Codanin1 binding site and suggesting alternative mechanisms. In the revised Discussion, we added a paragraph on alternative pathogenic models, p. 29, l. 702-713:
"Our study centers on the CDIN1-binding C-terminus, however, most CDA-I-associated CDAN1 mutations lie elsewhere and probably act through alternative mechanisms. Mutations such as P672L and F868I in the LOBE2 (452-798 amino acid residue) and F868I in the coiled-coil (841-1000 amino acid residue) domains may disturb Codanin1 homodimerization and higher-order complex assembly, directly affecting ASF1 sequestration (Jeong, T. K. et al. Nature Comm. 2025). Other mutant variants may also interfere with ASF1 sequestration, nuclear targeting, or chromatin-remodeling functions, while destabilizing mutations may induce misfolding and proteasomal degradation. Moreover, CDA-I-associated mutations, such as R714W and R1042W, might compromise the interaction between Codanin1 and ASF1 (Ask, Jasencakova et al. 2012). Collectively, the complementary alternative pathogenic mechanisms associated with Codanin1 mutations in distal regions and mutations in CDIN1‑binding C-terminus of Codanin1 may contribute to erythroid dysfunction in CDA-I."
Reviewer #3 – 4. ____Contradictory Functional Models:
Ask et al. (EMBO J, 2012) reported that Codanin-1 depletion increases nuclear ASF1 and accelerates DNA replication. This contrasts with the current hypothesis that disruption of the Codanin-1/CDIN1 complex reduces nuclear ASF1.
The authors should attempt to reconcile this apparent contradiction, possibly by proposing a context-specific or dual-function model for Codanin-1 in histone trafficking.
3.4 Response – ____Clarified explanation of hypothetical functional model
We thank the reviewer for raising this point, which improved the clarity of our work. There is no real discrepancy between Ask et al. and our findings; both agree that Codanin1 restrains ASF1 in the cytoplasm. Ask et al. examined the complete loss of Codanin1, which abolishes cytoplasmic ASF1 sequestration and thus leads to maximal nuclear accumulation. We suggest the CDA-I-associated mutations selectively disrupt the CDIN1-Codanin1 interface, releasing ASF1 from the cytoplasm into the nucleus.
To enhance clarity, we now state in the legend of Figure 4 describing the hypothesis (p. 31, l. 752-753): "…CDA-I-associated mutations prevent CDIN1-Codanin1 complex formation, thus prevent ASF1 sequestration to cytoplasm; ASF1 remains accumulated in nucleus."
Reviewer #3 – 5. ____Conclusions and Claims:
The proposed model of CDA I pathogenesis (Fig. 4) is plausible but not yet fully supported by the available data. The authors suggest that disruption of the Codanin-1/CDIN1 interaction leads to nuclear histone depletion, but this has not been experimentally confirmed.
Claims about the general pathogenesis of CDA I should be clearly qualified as hypothetical and applicable to a subset of mutations. The presence and localization of ASF1 in the nucleus following disruption of the Codanin-1/CDIN1 complex should be tested experimentally.
3.5 Response – __Tempered ____conclusions and claims: __
We thank the reviewer for underscoring the need to temper our conclusions and to distinguish hypotheses from available results. We fully agree that our Fig. 4 model—linking disruption of the Codanin1-CDIN1 interface to nuclear histone imbalance—remains a working hypothesis, currently supported by indirect biochemical and structural data.
Accordingly, we have:
Revised the text to explicitly state that this model is hypothetical and pertains to a subset of CDA-I-associated CDAN1 mutations. Specifically, we
Added to the last paragraph of the section Functional implications of CDA-I-related mutations in Discussion (p. 31, l. 744-749): “In considering functional implications of our findings within available data, it is essential to qualify that mechanistic claims regarding the general pathogenesis of CDA-I remain hypothetical and are restricted to a specific subset of mutations. Furthermore, direct experimental validation, such as immunolocalization or live-cell imaging, to assess ASF1’s nuclear presence and distribution following disruption of the CDIN1-Codanin1 complex is required to substantiate the proposed model.”
__Reviewer #3 – 6.____Broader Mutation Analysis and ASF1 Localization: __
To strengthen the link between Codanin-1/CDIN1 disruption and disease pathogenesis, it would be important to test the effects of additional CDAN1 mutations, including those outside the C-terminal region. Similarly, the impact on ASF1 nuclear concentration and localization should be directly assessed. These experiments would significantly bolster the central hypothesis. If feasible, they should be pursued or at least acknowledged as important future directions.
3.6 Response – Broader mutation analysis and ASF1 localization in future directions
We thank Reviewer #3 for emphasizing the value of a broader mutation survey and direct ASF1 localization studies. As noted above, our current manuscript is centered on delineating the molecular architecture of the CDIN1-Codanin1Cterm core interface; comprehensive mutational analyses outside the C-terminal binding region and ASF1-dependent functional assays will be critical to extend these findings but fall beyond the scope of the present work and will be the objective of our following studies. To address the reviewer’s concern, we have:
Expanded the Future Directions section to specify that additional CDA-I-linked CDAN1 variants, including non-C-terminal mutations, and quantitative assessments of ASF1 nuclear localization will be the subject of ongoing and planned investigations. Specifically, we added (p. 32, l. 776-778):” In future work, additional Codanin1 mutations, including those outside the C-terminal region, should be evaluated to determine how the mutations affect ASF1’s nuclear concentration and subcellular localization.”
Emphasized the need for complementary in vivo validation in erythroblast models to confirm whether the disturbance of CDIN1-Codanin1 binding recapitulates CDA-I phenotypes. We acknowledged the need for cell-line studies in future work within the Future research directions of Discussion (p. 32, l. 774-776): “Finally, follow-up research utilizing erythroblast model cell lines must be conducted to determine if specific mutations that disrupt CDIN1-Codanin1 binding, also affect ASF1 localization and cause a phenotype typical of CDA-I.” We believe these changes more precisely delimit the scope and significance of the current study while laying out a clear roadmap for the essential follow-up experiments.
Reviewer #3 – 7. ____Rigor and Presentation and Cross-commenting
__Minor Comments: __
• Methods and Reproducibility:
The experimental methods are well described, and the results appear reproducible.
• Presentation:
The text and figures are clear and well organized.
Referee Cross-commenting
I agree with reviewer 1 that the paper presents detailed structure study of Codanin-1 and CDIN1 protein. However, as reviewer 2 claims functional studies are missing and therefore the hypothesis regarding the pahtogenesis of CDAI is speculaltive especially with no studies regarding ASF1.
3____.7 Response ____–____ Rigor and Presentation and Cross-commenting:
We thank the reviewers for their positive appraisal of our results' reproducibility, presentation, and method descriptions. We also appreciate the cross-comment that, while our structural analysis of the CDIN1-Codanin1 complex is thorough, functional validation, particularly regarding ASF1, remains to be addressed.
As outlined above, we have revised the manuscript to:
__Reviewer #3 –____ Significance: __
Nature and Significance of the Advance:
This study extends prior work (e.g., Swickley et al., BMC Mol Cell Biol 2020; Shroff et al., Biochem J 2020) on Codanin-1/CDIN1 interaction by applying high-resolution biophysical techniques to identify mutations that disrupt this complex. It provides a plausible cellular mechanism by which specific mutations may lead to CDA I through impaired histone trafficking.
Nevertheless, key question remains: How do mutations outside the Codanin-1 C-terminus contribute to the pathology?
3.8 Response – Significance:
We revised the text to clarify how mutations beyond the C-terminus may contribute to CDA-I pathogenesis and present the significance of our current structural analyses, biophysical characterizations, and molecular insights as a foundation for future research (please refer to Response 3.6). __Audience: __
Molecular and cellular biologists investigating nuclear-cytoplasmic trafficking mechanisms
Pediatric hematologist with over 20 years of research experience in CDA I, including the initial identification of CDAN1 and the elucidation of Codanin-1's role in embryonic erythropoiesis. Not a specialist in the biophysical techniques used in this study.
References
Ask, K., Z. Jasencakova, P. Menard, Y. Feng, G. Almouzni and A. Groth (2012). "Codanin-1, mutated in the anaemic disease CDAI, regulates Asf1 function in S-phase histone supply." The EMBO Journal 31(8): 2013–2023.
Jeong, T.-K., R. C. M. Frater, J. Yoon, A. Groth and J.-J. Song (2025). "CODANIN-1 sequesters ASF1 by using a histone H3 mimic helix to regulate the histone supply." Nature Communications 16(1): 2181.
Sedor, S. F. and S. Shao (2025). "Mechanism of ASF1 engagement by CDAN1." Nature Communications 16(1): 2599.
Swickley, G., Y. Bloch, L. Malka, A. Meiri, S. Noy-Lotan, A. Yanai, H. Tamary and B. Motro (2020). "Characterization of the interactions between Codanin-1 and C15Orf41, two proteins implicated in congenital dyserythropoietic anemia type I disease." Molecular and Cell Biology 21(1).
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Congenital Dyserythropoietic Anemia type I (CDA I) is an autosomal recessive disorder characterized by ineffective erythropoiesis and distinctive nuclear morphology ("Swiss cheese" heterochromatin) in erythroblasts. CDA I is caused by mutations in CDAN1 and CDIN1. Codanin-1, encoded by CDAN1, is part of the cytosolic ASF1-H3.1-H4-Importin-4 complex, which regulates histone trafficking to the nucleus. CDIN1 has been shown to bind the C-terminal domain of Codanin-1, but until now, pathogenic mutations had not been directly linked to the disruption of this interaction. In this study, the authors used biophysical techniques to characterize the interaction between Codanin-1's C-terminal region (residues 1005-1227) and CDIN1, demonstrating high-affinity, equimolar binding. HDX-MS identified interaction hotspots, and disease-associated mutations in these regions disrupted complex formation. The authors propose that such disruption prevents ASF1 sequestration in the cytoplasm, thereby reducing nuclear histone levels and contributing to the chromatin abnormalities seen in CDA I.
Major Comments:
Minor Comments:
Referee Cross-commenting
I agree with reviewer 1 that the paper present detailed strucutre study of Codann-1 and CDIN1 protein. However, as reviewer 2 claims functional studies are missing and therefore the hypothesis regarding the pahtogenesis of CDAI is speculaltive especially with no studies regarding ASF1.
Nature and Significance of the Advance:
This study extends prior work (e.g., Swickley et al., BMC Mol Cell Biol 2020; Shroff et al., Biochem J 2020) on Codanin-1/CDIN1 interaction by applying high-resolution biophysical techniques to identify mutations that disrupt this complex. It provides a plausible cellular mechanism by which specific mutations may lead to CDA I through impaired histone trafficking. Nevertheless, key question remains: How do mutations outside the Codanin-1 C-terminus contribute to the pathology?
Audience:
Molecular and cellular biologists investigating nuclear-cytoplasmic trafficking mechanisms Hematologists and geneticists studying rare red cell disorders Clinicians managing CDA I patients and researchers exploring targeted therapies
Reviewer Expertise:
Pediatric hematologist with over 20 years of research experience in CDA I, including the initial identification of CDAN1 and the elucidation of Codanin-1's role in embryonic erythropoiesis. Not a specialist in the biophysical techniques used in this study
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript presents structural and biochemical characterization of the interaction between CDIN1 and the C-terminal domain of Codanin1, shedding light on a complex implicated in Congenital Dyserythropoietic Anemia Type I (CDA-I). While the authors provide valuable structural insights and identify disease-associated mutations that impair CDIN1-Codanin1 binding, I think several important concerns should be addressed to strengthen both the mechanistic claims and their functional relevance.
Contradiction Between Stoichiometry Models:
The authors propose that CDIN1 and Codanin1Cterm primarily form a heterodimer in vitro. However, this appears to contradict previous reports indicating a tetra-heteromeric arrangement. Additionally, while CDIN1 homodimerize seems confusing to me, do the authors suggest it is stable without Codanin1? This seems contrary to findings that CDIN1 is unstable in the absence of Codanin1(Sedor, S.F., Shao, S. nature comm 2025, Swickley, G., Bloch, Y., Malka, L. et al 2020 BMC Mol and Cell Biol). These inconsistencies raise concerns about whether the observed stoichiometries are physiologically relevant or artifacts of in vitro reconstitution, especially since full-length Codanin1 was not studied.
Unvalidated Functional Claims:
The manuscript identifies several CDA-I-associated mutations that disrupt CDIN1-Codanin1 interaction. However, the authors do not test how these mutations affect the biological function of the complex, particularly its role in ASF1 sequestration or histone trafficking. Given the central importance of this axis in their disease model, functional validation (e.g., ASF1 localization, histone deposition assays) is necessary to support these mechanistic conclusions.
Speculative and Potentially Contradictory Model:
The proposed model suggests that CDIN1 competes with ASF1 for Codanin1 binding, thereby indirectly promoting histone delivery to the nucleus. However, emerging data indicate that Codanin1, CDIN1, and ASF1 can form a stable ternary complex, calling into question this competitive binding hypothesis (Sedor, S.F., Shao, S. nature comm 2025). The authors do not acknowledge or discuss these findings, and the model in its current form may therefore be oversimplified or inaccurate.
Overall, the study adds to our structural understanding of CDIN1 and Codanin1 interactions, but the functional interpretations are currently speculative, and in some cases in conflict with existing literature. The manuscript would benefit significantly from addressing these discrepancies, incorporating relevant data on ASF1, and clarifying whether the observed assemblies reflect physiological complexes.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This is a rigorous biophysical characterization of a protein-protein interaction relevant to CDA-1 disease. The two proteins were purified in an E. coli host but CD and DLS was performed to ensure that the purified protein is well folded. An impressive native protein EMSA was used to show a 1:1 complex. While common for protein-nucleic acid complexes, EMSAs are much more challenging with protein complexes. A higher-running complex, likely a heterotetramer was implied at higher protein concentrations. These results were supported with SEC-MALS analysis and analytic ultracentrifugation analysis. Thermophoresis and ITC were used to report a nanomolar affinity of the proteins for each other. SEC-SAXS supported the conclusions about stoichiometry and composition inferred from the earlier methods and suggested that the dimerization interface comes from CDIN1. Next HDX-MS was used to identify putative interface residues, which were then mutated in each of the proteins and assessed for binding using coimmunoprecipitation. This study uses at least 10 orthogonal biophysical and/or biochemical methodologies to characterize an important protein-protein interaction and the analysis is clear and so is the writing. I couldn't (reading it once) find any grammatical or other errors in the text or figures. This manuscript is top-quality and suitable for publication.
Such detailed structural and mechanistic studies are greatly lacking in many clinical conditions for which mutations are known (unless they cause cancer, neurodegenerative disease, and so on). We need more such studies on disease topics! This study will be of interest for the hematologic diseases community.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reply to the Reviewers
I would like to thank the reviewers for their comments and interest in the manuscript and the study.
Reviewer #1
1. I would assume that there are RNA-seq and/or ChIP-seq data out there produced after knockdown of one or more of these DBPs that show directional positioning.
The directional positioning of CTCF-binding sites at chromatin interaction sites was analyzed by CRISPR experiment (Guo Y et al. Cell 2015). We found that the machine learning and statistical analysis showed the same directional bias of CTCF-binding motif sequence and RAD21-binding motif sequence at chromatin interaction sites as the experimental analysis of Guo Y et al. (lines 229-253, Figure 3b, c, d and Table 1). Since CTCF is involved in different biological functions (Braccioli L et al. Essays Biochem. 2019 ResearchGate webpage), the directional bias of binding sites may be reduced in all binding sites including those at chromatin interaction sites (lines 68-73). In our study, we investigated the DNA-binding sites of proteins using the ChIP-seq data of DNA-binding proteins and DNase-seq data. We also confirmed that the DNA-binding sites of SMC3 and RAD21, which tend to be found in chromatin loops with CTCF, also showed the same directional bias as CTCF by the computational analysis.
__2. Figure 6 should be expanded to incorporate analysis of DBPs not overlapping CTCF/cohesin in chromatin interaction data that is important and potentially more interesting than the simple DBPs enrichment reported in the present form of the figure. __
Following the reviewer's advice, I performed the same analysis with the DNA-binding sites that do no overlap with the DNA-binding sites of CTCF and cohesin (RAD21 and SMC3) (Fig. 6 and Supplementary Fig. 4). The result showed the same tendency in the distribution of DNA-binding sites. The height of a peak on the graph became lower for some DNA-binding proteins after removing the DNA-binding sites that overlapped with those of CTCF and cohesin. I have added the following sentence on lines 435 and 829: For the insulator-associated DBPs other than CTCF, RAD21, and SMC3, the DNA-binding sites that do not overlap with those of CTCF, RND21, and SMC3 were used to examine their distribution around interaction sites.
3. Critically, I would like to see use of Micro-C/Hi-C data and ChIP-seq from these factors, where insulation scores around their directionally-bound sites show some sort of an effect like that presumed by the authors - and many such datasets are publicly-available and can be put to good use here.
As suggested by the reviewer, I have added the insulator scores and boundary sites from the 4D nucleome data portal as tracks in the UCSC genome browser. The insulator scores seem to correspond to some extent to the H3K27me3 histone marks from ChIP-seq (Fig. 4a and Supplementary Fig. 3). We found that the DNA-binding sites of the insulator-associated DBPs were statistically overrepresented in the 5 kb boundary sites more than other DBPs (Fig. 4d). The direction of DNA-binding sites on the genome can be shown with different colors (e.g. red and green), but the directionality of insulator-associated DNA-binding sites is their overall tendency, and it may be difficult to notice the directionality from each binding site because the directionality may be weaker than that of CTCF, RAD21, and SMC3 as shown in Table 1 and Supplementary Table 2. We also observed the directional biases of CTCF, RAD21, and SMC3 by using Micro-C chromatin interaction data as we estimated, but the directionality was more apparent to distinguish the differences between the four directions of FR, RF, FF, and RR using CTCF-mediated ChIA-pet chromatin interaction data (lines 287 and 288).
I found that the CTCF binding sites examined by a wet experiment in the previous study may not always overlap with the boundary sites of chromatin interactions from Micro-C assay (Guo Y et al. *Cell* 2015). The chromatin interaction data do not include all interactions due to the high sequencing cost of the assay, and include less long-range interactions due to distance bias. The number of the boundary sites may be smaller than that of CTCF binding sites acting as insulators and/or some of the CTCF binding sites may not be locate in the boundary sites. It may be difficult for the boundary location algorithm to identify a short boundary location. Due to the limitations of the chromatin interaction data, I planned to search for insulator-associated DNA-binding proteins without using chromatin interaction data in this study.
I discussed other causes in lines 614-622: Another reason for the difference may be that boundary sites are more closely associated with topologically associated domains (TADs) of chromosome than are insulator sites. Boundary sites are regions identified based on the separation of numerous chromatin interactions. On the other hand, we found that the multiple DNA-binding sites of insulator-associated DNA-binding proteins were located close to each other at insulator sites and were associated with distinct nested and focal chromatin interactions, as reported by Micro-C assay. These interactions may be transient and relatively weak, such as tissue/cell type, conditional or lineage-specific interactions.
Furthermore, I have added the statistical summary of the analysis in lines 372-395 as follows: Overall, among 20,837 DNA-binding sites of the 97 insulator-associated proteins found at insulator sites identified by H3K27me3 histone modification marks (type 1 insulator sites), 1,315 (6%) overlapped with 264 of 17,126 5kb long boundary sites, and 6,137 (29%) overlapped with 784 of 17,126 25kb long boundary sites in HFF cells. Among 5,205 DNA-binding sites of the 97 insulator-associated DNA-binding proteins found at insulator sites identified by H3K27me3 histone modification marks and transcribed regions (type 2 insulator sites), 383 (7%) overlapped with 74 of 17,126 5-kb long boundary sites, 1,901 (37%) overlapped with 306 of 17,126 25-kb long boundary sites. Although CTCF-binding sites separate active and repressive domains, the limited number of DNA-binding sites of insulator-associated proteins found at type 1 and 2 insulator sites overlapped boundary sites identified by chromatin interaction data. Furthermore, by analyzing the regulatory regions of genes, the DNA-binding sites of the 97 insulator-associated DNA-binding proteins were found (1) at the type 1 insulator sites (based on H3K27me3 marks) in the regulatory regions of 3,170 genes, (2) at the type 2 insulator sites (based on H3K27me3 marks and gene expression levels) in the regulatory regions of 1,044 genes, and (3) at insulator sites as boundary sites identified by chromatin interaction data in the regulatory regions of 6,275 genes. The boundary sites showed the highest number of overlaps with the DNA-binding sites. Comparing the insulator sites identified by (1) and (3), 1,212 (38%) genes have both types of insulator sites. Comparing the insulator sites between (2) and (3), 389 (37%) genes have both types of insulator sites. From the comparison of insulator and boundary sites, we found that (1) or (2) types of insulator sites overlapped or were close to boundary sites identified by chromatin interaction data.
4. The suggested alternative transcripts function, also highlighted in the manuscripts abstract, is only supported by visual inspection of a few cases for several putative DBPs. I believe this is insufficient to support what looks like one of the major claims of the paper when reading the abstract, and a more quantitative and genome-wide analysis must be adopted, although the authors mention it as just an 'observation'.
According to the reviewer's comment, I performed the genome-wide analysis of alternative transcripts where the DNA-binding sites of insulator-associated proteins are located near splicing sites. The DNA-binding sites of insulator-associated DNA-binding proteins were found within 200 bp centered on splice sites more significantly than the other DNA-binding proteins (Fig. 4e and Table 2). I have added the following sentences on lines 405 - 412: We performed the statistical test to estimate the enrichment of insulator-associated DNA-binding sites compared to the other DNA-binding proteins, and found that the insulator-associated DNA-binding sites were significantly more abundant at splice sites than the DNA-binding sites of the other proteins (Fig 4e and Table 2; Mann‒Whitney U test, p value 5. Figure 1 serves no purpose in my opinion and can be removed, while figures can generally be improved (e.g., the browser screenshots in Figs 4 and 5) for interpretability from readers outside the immediate research field.
I believe that the Figure 1 would help researchers in other fields who are not familiar with biological phenomena and functions to understand the study. More explanation has been included in the Figures and legends of Figs. 4 and 5 to help readers outside the immediate research field understand the figures.
6. Similarly, the text is rather convoluted at places and should be re-approached with more clarity for less specialized readers in mind.
Reviewer #2's comments would be related to this comment. I have introduced a more detailed explanation of the method in the Results section, as shown in the responses to Reviewer #2's comments.
Reviewer #2
1. Introduction, line 95: CTCF appears two times, it seems redundant.
On lines 91-93, I deleted the latter CTCF from the sentence "We examine the directional bias of DNA-binding sites of CTCF and insulator-associated DBPs, including those of known DBPs such as RAD21 and SMC3".
2. Introduction, lines 99-103: Please stress better the novelty of the work. What is the main focus? The new identified DPBs or their binding sites? What are the "novel structural and functional roles of DBPs" mentioned?
Although CTCF is known to be the main insulator protein in vertebrates, we found that 97 DNA-binding proteins including CTCF and cohesin are associated with insulator sites by modifying and developing a machine learning method to search for insulator-associated DNA-binding proteins. Most of the insulator-associated DNA-binding proteins showed the directional bias of DNA-binding motifs, suggesting that the directional bias is associated with the insulator.
I have added the sentence in lines 96-99 as follows: Furthermore, statistical testing the contribution scores between the directional and non-directional DNA-binding sites of insulator-associated DBPs revealed that the directional sites contributed more significantly to the prediction of gene expression levels than the non-directional sites. I have revised the statement in lines 101-110 as follows: To validate these findings, we demonstrate that the DNA-binding sites of the identified insulator-associated DBPs are located within potential insulator sites, and some of the DNA-binding sites in the insulator site are found without the nearby DNA-binding sites of CTCF and cohesin. Homologous and heterologous insulator-insulator pairing interactions are orientation-dependent, as suggested by the insulator-pairing model based on experimental analysis in flies. Our method and analyses contribute to the identification of insulator- and chromatin-associated DNA-binding sites that influence EPIs and reveal novel functional roles and molecular mechanisms of DBPs associated with transcriptional condensation, phase separation and transcriptional regulation.
3. Results, line 111: How do the SNPs come into the procedure? From the figures it seems the input is ChIP-seq peaks of DNBPs around the TSS.
On lines 121-124, to explain the procedure for the SNP of an eQTL, I have added the sentence in the Methods: "If a DNA-binding site was located within a 100-bp region around a single-nucleotide polymorphism (SNP) of an eQTL, we assumed that the DNA-binding proteins regulated the expression of the transcript corresponding to the eQTL".
4. Again, are those SNPs coming from the different cell lines? Or are they from individuals w.r.t some reference genome? I suggest a general restructuring of this part to let the reader understand more easily. One option could be simplifying the details here or alternatively including all the necessary details.
On line 119, I have included the explanation of the eQTL dataset of GTEx v8 as follows: " The eQTL data were derived from the GTEx v8 dataset, after quality control, consisting of 838 donors and 17,382 samples from 52 tissues and two cell lines". On lines 681 and 865, I have added the filename of the eQTL data "(GTEx_Analysis_v8_eQTL.tar)".
5. Figure 1: panel a and b are misleading. Is the matrix in panel a equivalent to the matrix in panel b? If not please clarify why. Maybe in b it is included the info about the SNPs? And if yes, again, what is then difference with a.
The reviewer would mention Figure 2, not Figure 1. If so, the matrices in panels a and b in Figure 2 are equivalent. I have shown it in the figure: The same figure in panel a is rotated 90 degrees to the right. The green boxes in the matrix show the regions with the ChIP-seq peak of a DNA-binding protein overlapping with a SNP of an eQTL. I used eQTL data to associate a gene with a ChIP-seq peak that was more than 2 kb upstream and 1 kb downstream of a transcriptional start site of a gene. For each gene, the matrix was produced and the gene expression levels in cells were learned and predicted using the deep learning method. I have added the following sentences to explain the method in lines 133 - 139: Through the training, the tool learned to select the binding sites of DNA-binding proteins from ChIP-seq assays that were suitable for predicting gene expression levels in the cell types. The binding sites of a DNA-binding protein tend to be observed in common across multiple cell and tissue types. Therefore, ChIP-seq data and eQTL data in different cell and tissue types were used as input data for learning, and then the tool selected the data suitable for predicting gene expression levels in the cell types, even if the data were not obtained from the same cell types.
6. Line 386-388: could the author investigate in more detail this observation? Does it mean that loops driven by other DBPs independent of the known CTCF/Cohesin? Could the author provide examples of chromatin structural data e.g. MicroC?
As suggested by the reviewer, to help readers understand the observation, I have added Supplementary Fig. S4c to show the distribution of DNA-binding sites of "CTCF, RAD21, and SMC3" and "BACH2, FOS, ATF3, NFE2, and MAFK" around chromatin interaction sites. I have modified the following sentence to indicate the figure on line 501: Although a DNA-binding-site distribution pattern around chromatin interaction sites similar to those of CTCF, RAD21, and SMC3 was observed for DBPs such as BACH2, FOS, ATF3, NFE2, and MAFK, less than 1% of the DNA-binding sites of the latter set of DBPs colocalized with CTCF, RAD21, or SMC3 in a single bin (Fig. S4c).
In Aljahani A et al. *Nature Communications* 2022, we find that depletion of cohesin causes a subtle reduction in longer-range enhancer-promoter interactions and that CTCF depletion can cause rewiring of regulatory contacts. Together, our data show that loop extrusion is not essential for enhancer-promoter interactions, but contributes to their robustness and specificity and to precise regulation of gene expression. Goel VY et al. *Nature Genetics* 2023 mentioned in the abstract: Microcompartments frequently connect enhancers and promoters and though loss of loop extrusion and inhibition of transcription disrupts some microcompartments, most are largely unaffected. These results suggested that chromatin loops can be driven by other DBPs independent of the known CTCF/Cohesin.
I added the following sentence on lines 569-577: The depletion of cohesin causes a subtle reduction in longer-range enhancer-promoter interactions and that CTCF depletion can cause rewiring of regulatory contacts. Another group reported that enhancer-promoter interactions and transcription are largely maintained upon depletion of CTCF, cohesin, WAPL or YY1. Instead, cohesin depletion decreased transcription factor binding to chromatin. Thus, cohesin may allow transcription factors to find and bind their targets more efficiently. Furthermore, the loop extrusion is not essential for enhancer-promoter interactions, but contributes to their robustness and specificity and to precise regulation of gene expression.
FOXA1 pioneer factor functions as an initial chromatin-binding and chromatin-remodeling factor and has been reported to form biomolecular condensates (Ji D et al. *Molecular Cell* 2024). CTCF have also found to form transcriptional condensate and phase separation (Lee R et al. *Nucleic acids research* 2022). FOS was found to be an insulator-associated DNA-binding protein in this study and is potentially involved in chromatin remodeling, transcription condensation, and phase separation with the other factors such as BACH2, ATF3, NFE2 and MAFK. I have added the following sentence on line 556: FOXA1 pioneer factor functions as an initial chromatin-binding and chromatin-remodeling factor and has been reported to form biomolecular condensates.
7. In general, how the presented results are related to some models of chromatin architecture, e.g. loop extrusion, in which it is integrated convergent CTCF binding sites?
Goel VY et al. Nature Genetics 2023 identified highly nested and focal interactions through region capture Micro-C, which resemble fine-scale compartmental interactions and are termed microcompartments. In the section titled "Most microcompartments are robust to loss of loop extrusion," the researchers noted that a small proportion of interactions between CTCF and cohesin-bound sites exhibited significant reductions in strength when cohesin was depleted. In contrast, the majority of microcompartmental interactions remained largely unchanged under cohesin depletion. Our findings indicate that most P-P and E-P interactions, aside from a few CTCF and cohesin-bound enhancers and promoters, are likely facilitated by a compartmentalization mechanism that differs from loop extrusion. We suggest that nested, multiway, and focal microcompartments correspond to small, discrete A-compartments that arise through a compartmentalization process, potentially influenced by factors upstream of RNA Pol II initiation, such as transcription factors, co-factors, or active chromatin states. It follows that if active chromatin regions at microcompartment anchors exhibit selective "stickiness" with one another, they will tend to co-segregate, leading to the development of nested, focal interactions. This microphase separation, driven by preferential interactions among active loci within a block copolymer, may account for the striking interaction patterns we observe.
The authors of the paper proposed several mechanisms potentially involved in microcompartments. These mechanisms may be involved in looping with insulator function. Another group reported that enhancer-promoter interactions and transcription are largely maintained upon depletion of CTCF, cohesin, WAPL or YY1. Instead, cohesin depletion decreased transcription factor binding to chromatin. Thus, cohesin may allow transcription factors to find and bind their targets more efficiently (Hsieh TS et al. *Nature Genetics* 2022). Among the identified insulator-associated DNA-binding proteins, Maz and MyoD1 form loops without CTCF (Xiao T et al. *Proc Natl Acad Sci USA* 2021 ; Ortabozkoyun H et al. *Nature genetics* 2022 ; Wang R et al. *Nature communications* 2022). I have added the following sentences on lines 571-575: Another group reported that enhancer-promoter interactions and transcription are largely maintained upon depletion of CTCF, cohesin, WAPL or YY1. Instead, cohesin depletion decreased transcription factor binding to chromatin. Thus, cohesin may allow transcription factors to find and bind their targets more efficiently. I have included the following explanation on lines 582-584: Maz and MyoD1 among the identified insulator-associated DNA-binding proteins form loops without CTCF.
As for the directionality of CTCF, if chromatin loop anchors have some structural conformation, as shown in the paper entitled "The structural basis for cohesin-CTCF-anchored loops" (Li Y et al. *Nature* 2020), directional DNA binding would occur similarly to CTCF binding sites. Moreover, cohesin complexes that interact with convergent CTCF sites, that is, the N-terminus of CTCF, might be protected from WAPL, but those that interact with divergent CTCF sites, that is, the C-terminus of CTCF, might not be protected from WAPL, which could release cohesin from chromatin and thus disrupt cohesin-mediated chromatin loops (Davidson IF et al. *Nature Reviews Molecular Cell Biology* 2021). Regarding loop extrusion, the 'loop extrusion' hypothesis is motivated by in vitro observations. The experiment in yeast, in which cohesin variants that are unable to extrude DNA loops but retain the ability to topologically entrap DNA, suggested that in vivo chromatin loops are formed independently of loop extrusion. Instead, transcription promotes loop formation and acts as an extrinsic motor that extends these loops and defines their final positions (Guerin TM et al. *EMBO Journal* 2024). I have added the following sentences on lines 543-547: Cohesin complexes that interact with convergent CTCF sites, that is, the N-terminus of CTCF, might be protected from WAPL, but those that interact with divergent CTCF sites, that is, the C-terminus of CTCF, might not be protected from WAPL, which could release cohesin from chromatin and thus disrupt cohesin-mediated chromatin loops. I have included the following sentences on lines 577-582: The 'loop extrusion' hypothesis is motivated by in vitro observations. The experiment in yeast, in which cohesin variants that are unable to extrude DNA loops but retain the ability to topologically entrap DNA, suggested that in vivo chromatin loops are formed independently of loop extrusion. Instead, transcription promotes loop formation and acts as an extrinsic motor that extends these loops and defines their final positions.
Another model for the regulation of gene expression by insulators is the boundary-pairing (insulator-pairing) model (Bing X et al. *Elife* 2024) (Ke W et al. *Elife* 2024) (Fujioka M et al. *PLoS Genetics* 2016). Molecules bound to insulators physically pair with their partners, either head-to-head or head-to-tail, with different degrees of specificity at the termini of TADs in flies. Although the experiments do not reveal how partners find each other, the mechanism unlikely requires loop extrusion. Homologous and heterologous insulator-insulator pairing interactions are central to the architectural functions of insulators. The manner of insulator-insulator interactions is orientation-dependent. I have summarized the model on lines 559-567: Other types of chromatin regulation are also expected to be related to the structural interactions of molecules. As the boundary-pairing (insulator-pairing) model, molecules bound to insulators physically pair with their partners, either head-to-head or head-to-tail, with different degrees of specificity at the termini of TADs in flies (Fig. 7). Although the experiments do not reveal how partners find each other, the mechanism unlikely requires loop extrusion. Homologous and heterologous insulator-insulator pairing interactions are central to the architectural functions of insulators. The manner of insulator-insulator interactions is orientation-dependent.
8. Do the authors think that the identified DBPs could work in that way as well?
The boundary-pairing (insulator-pairing) model would be applied to the insulator-associated DNA-binding proteins other than CTCF and cohesin that are involved in the loop extrusion mechanism (Bing X et al. Elife 2024) (Ke W et al. Elife 2024) (Fujioka M et al. PLoS Genetics 2016).
Liquid-liquid phase separation was shown to occur through CTCF-mediated chromatin loops and to act as an insulator (Lee, R et al. *Nucleic Acids Research* 2022). Among the identified insulator-associated DNA-binding proteins, CEBPA has been found to form hubs that colocalize with transcriptional co-activators in a native cell context, which is associated with transcriptional condensate and phase separation (Christou-Kent M et al. *Cell Reports* 2023). The proposed microcompartment mechanisms are also associated with phase separation. Thus, the same or similar mechanisms are potentially associated with the insulator function of the identified DNA-binding proteins. I have included the following information on line 554: CEBPA in the identified insulator-associated DNA-binding proteins was also reported to be involved in transcriptional condensates and phase separation.
9. Also, can the authors comment about the mechanisms those newly identified DBPs mediate contacts by active processes or equilibrium processes?
Snead WT et al. Molecular Cell 2019 mentioned that protein post-transcriptional modifications (PTMs) facilitate the control of molecular valency and strength of protein-protein interactions. O-GlcNAcylation as a PTM inhibits CTCF binding to chromatin (Tang X et al. Nature Communications 2024). I found that the identified insulator-associated DNA-binding proteins tend to form a cluster at potential insulator sites (Supplementary Fig. 2d). These proteins may interact and actively regulate chromatin interactions, transcriptional condensation, and phase separation by PTMs. I have added the following explanation on lines 584-590: Furthermore, protein post-transcriptional modifications (PTMs) facilitate control over the molecular valency and strength of protein-protein interactions. O-GlcNAcylation as a PTM inhibits CTCF binding to chromatin. We found that the identified insulator-associated DNA-binding proteins tend to form a cluster at potential insulator sites (Fig. 4f and Supplementary Fig. 3c). These proteins may interact and actively regulate chromatin interactions, transcriptional condensation, and phase separation through PTMs.
10. Can the author provide some real examples along with published structural data (e.g. the mentioned micro-C data) to show the link between protein co-presence, directional bias and contact formation?
Structural molecular model of cohesin-CTCF-anchored loops has been published by Li Y et al. Nature 2020. The structural conformation of CTCF and cohesin in the loops would be the cause of the directional bias of CTCF binding sites, which I mentioned in lines 539 - 543 as follows: These results suggest that the directional bias of DNA-binding sites of insulator-associated DBPs may be involved in insulator function and chromatin regulation through structural interactions among DBPs, other proteins, DNAs, and RNAs. For example, the N-terminal amino acids of CTCF have been shown to interact with RAD21 in chromatin loops.
To investigate the principles underlying the architectural functions of insulator-insulator pairing interactions, two insulators, Homie and Nhomie, flanking the *Drosophila even skipped *locus were analyzed. Pairing interactions between the transgene Homie and the eve locus are directional. The head-to-head pairing between the transgene and endogenous Homie matches the pattern of activation (Fujioka M et al. *PLoS Genetics* 2016).
Reviewer #3
Major Comments:
1. Some of these TFs do not have specific direct binding to DNA (P300, Cohesin). Since the authors are using binding motifs in their analysis workflow, I would remove those from the analysis.
When a protein complex binds to DNA, one protein of the complex binds to the DNA directory, and the other proteins may not bind to DNA. However, the DNA motif sequence bound by the protein may be registered as the DNA-binding motif of all the proteins in the complex. The molecular structure of the complex of CTCF and Cohesin showed that both CTCF and Cohesin bind to DNA (Li Y et al. Nature 2020). I think there is a possibility that if the molecular structure of a protein complex becomes available, the previous recognition of the DNA-binding ability of a protein may be changed. Therefore, I searched the Pfam database for 99 insulator-associated DNA-binding proteins identified in this study. I found that 97 are registered as DNA-binding proteins and/or have a known DNA-binding domain, and EP300 and SIN3A do not directory bind to DNA, which was also checked by Google search. I have added the following explanation in line 257 to indicate direct and indirect DNA-binding proteins: Among 99 insulator-associated DBPs, EP300 and SIN3A do not directory interact with DNA, and thus 97 insulator-associated DBPs directory bind to DNA. I have updated the sentence in line 20 of the Abstract as follows: We discovered 97 directional and minor nondirectional motifs in human fibroblast cells that corresponded to 23 DBPs related to insulator function, CTCF, and/or other types of chromosomal transcriptional regulation reported in previous studies.
2. I am not sure if I understood correctly, by why do the authors consider enhancers spanning 2Mb (200 bins of 10Kb around eSNPs)? This seems wrong. Enhancers are relatively small regions (100bp to 1Kb) and only a very small subset form super enhancers.
As the reviewer mentioned, I recognize enhancers are relatively small regions. In the paper, I intended to examine further upstream and downstream of promoter regions where enhancers are found. Therefore, I have modified the sentence in lines 929 - 931 of the Fig. 2 legend as follows: Enhancer-gene regulatory interaction regions consist of 200 bins of 10 kbp between -1 Mbp and 1 Mbp region from TSS, not including promoter.
3. I think the H3K27me3 analysis was very good, but I would have liked to see also constitutive heterochromatin as well, so maybe repeat the analysis for H3K9me3.
Following the reviewer's advice, I have added the ChIP-seq data of H3K9me3 as a truck of the UCSC Genome Browser. The distribution of H3K9me3 signal was different from that of H3K27me3 in some regions. I also found the insulator-associated DNA-binding sites close to the edges of H3K9me3 regions and took some screenshots of the UCSC Genome Browser of the regions around the sites in Supplementary Fig. 3b. I have modified the following sentence on lines 974 - 976 in the legend of Fig. 4: a Distribution of histone modification marks H3K27me3 (green color) and H3K9me3 (turquoise color) and transcript levels (pink color) in upstream and downstream regions of a potential insulator site (light orange color). I have also added the following result on lines 356 - 360: The same analysis was performed using H3K9me3 marks, instead of H3K27me3 (Fig. S3b). We found that the distribution of H3K9me3 signal was different from that of H3K27me3 in some regions, and discovered the insulator-associated DNA-binding sites close to the edges of H3K9me3 regions (Fig. S3b).
4. I was not sure I understood the analysis in Figure 6. The binding site is with 500bp of the interaction site, but micro-C interactions are at best at 1Kb resolution. They say they chose the centre of the interaction site, but we don't know exactly where there is the actual interaction. Also, it is not clear what they measure. Is it the number of binding sites of a specific or multiple DBP insulator proteins at a specific distance from this midpoint that they recover in all chromatin loops? Maybe I am missing something. This analysis was not very clear.
The resolution of the Micro-C assay is considered to be 100 bp and above, as the human nucleome core particle contains 145 bp (and 193 bp with linker) of DNA. However, internucleosomal DNA is cleaved by endonuclease into fragments of multiples of 10 nucleotides (Pospelov VA et al. Nucleic Acids Research 1979). Highly nested focal interactions were observed (Goel VY et al. Nature Genetics 2023). Base pair resolution was reported using Micro Capture-C (Hua P et al. Nature 2021). Sub-kilobase (20 bp resolution) chromatin topology was reported using an MNase-based chromosome conformation capture (3C) approach (Aljahani A et al. Nature Communications 2022). On the other hand, Hi-C data was analyzed at 1 kb resolution. (Gu H et al. bioRxiv 2021). If the resolution of Micro-C interactions is at best at 1 kb, the binding sites of a DNA-binding protein will not show a peak around the center of the genomic locations of interaction edges. Each panel shows the number of binding sites of a specific DNA-binding protein at a specific distance from the midpoint of all chromatin interaction edges. I have modified and added the following sentences in lines 593-597: High-resolution chromatin interaction data from a Micro-C assay indicated that most of the predicted insulator-associated DBPs showed DNA-binding-site distribution peaks around chromatin interaction sites, suggesting that these DBPs are involved in chromatin interactions and that the chromatin interaction data has a high degree of resolution. Base pair resolution was reported using Micro Capture-C.
Minor Comments:
1. PIQ does not consider TF concentration. Other methods do that and show that TF concentration improves predictions (e.g., ____https://www.biorxiv.org/content/10.1101/2023.07.15.549134v2____or ____https://pubmed.ncbi.nlm.nih.gov/37486787____/). The authors should discuss how that would impact their results.
The directional bias of CTCF binding sites was identified by ChIA-pet interactions of CTCF binding sites. The analysis of the contribution scores of DNA-binding sites of proteins considering the binding sites of CTCF as an insulator showed the same tendency of directional bias of CTCF binding sites. In the analysis, to remove the false-positive prediction of DNA-binding sites, I used the binding sites that overlapped with a ChIP-seq peak of the DNA-binding protein. This result suggests that the DNA-binding sites of CTCF obtained by the current analysis have sufficient quality. Therefore, if the accuracy of prediction of DNA-binding sites is improved, although the number of DNA-binding sites may be different, the overall tendency of the directionality of DNA-binding sites will not change and the results of this study will not change significantly.
As for the first reference in the reviewer's comment, chromatin interaction data from Micro-C assay does not include all chromatin interactions in a cell or tissue, because it is expensive to cover all interactions. Therefore, it would be difficult to predict all chromatin interactions based on machine learning. As for the second reference in the reviewer's comment, pioneer factors such as FOXA are known to bind to closed chromatin regions, but transcription factors and DNA-binding proteins involved in chromatin interactions and insulators generally bind to open chromatin regions. The search for the DNA-binding motifs is not required in closed chromatin regions.
2. DeepLIFT is a good approach to interpret complex structures of CNN, but is not truly explainable AI. I think the authors should acknowledge this.
In the DeepLIFT paper, the authors explain that DeepLIFT is a method for decomposing the output prediction of a neural network on a specific input by backpropagating the contributions of all neurons in the network to every feature of the input (Shrikumar A et al. ICML 2017). DeepLIFT compares the activation of each neuron to its 'reference activation' and assigns contribution scores according to the difference. DeepLIFT calculates a metric to measure the difference between an input and the reference of the input.
Truly explainable AI would be able to find cause and reason, and to make choices and decisions like humans. DeepLIFT does not perform causal inferences. I did not use the term "Explainable AI" in our manuscript, but I briefly explained it in Discussion. I have added the following explanation in lines 623-628: AI (Artificial Intelligence) is considered as a black box, since the reason and cause of prediction are difficult to know. To solve this issue, tools and methods have been developed to know the reason and cause. These technologies are called Explainable AI. DeepLIFT is considered to be a tool for Explainable AI. However, DeepLIFT does not answer the reason and cause for a prediction. It calculates scores representing the contribution of the input data to the prediction.
Furthermore, to improve the readability of the manuscript, I have included the following explanation in lines 159-165: we computed DeepLIFT scores of the input data (i.e., each binding site of the ChIP-seq data of DNA-binding proteins) in the deep leaning analysis on gene expression levels. DeepLIFT compares the importance of each input for predicting gene expression levels to its 'reference or background level' and assigns contribution scores according to the difference. DeepLIFT calculates a metric to measure the difference between an input and the reference of the input.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
Osato and Hamada propose a systematic approach to identify DNA binding proteins that display directional binding. They used a modified Deep Learning method (DEcode) to investigate binding profiles of 1356 DBP from GTRD database at promoters (30 of 100bp bins around TSS) and enhancers (200 bins of 10Kb around eSNPs) and use this to predict expression of 25,071 genes in Fibroblasts, Monocytes, HMEC and NPC. This method achieves a good prediction power (Spearman correlation between predicted and actual expression of 0.74). They then use PIQ, and overlap predicted binding sites with actual ChIP-seq data to investigate the motifs of TFs that are controlling gene expression. They find 99 insulator proteins showing either a specific directional bias or minor non-directional bias, corresponding to 23 DBP previously reported to have insulator function. Of the 23 proteins they identify as regulating enhancer promoter interactions, 13 are associated with CTCF. They also show that there are significantly more insulator proteins binding sites at borders of polycomb domains, transcriptionally active or boundary regions based on chromatin interactions than other proteins.
Major Comments:
Minor comments:
Referee Cross-Commenting
I would like to mention that I agree with the comments of reviewers 1 and 2.
General assessment:
This is the first study to my knowledge that attempts to use Deep Learning to identify insulators and directional biases in binding. One of the limitations is that no additional methods were used to show that these DBP have directional binding bias. It is not necessarily to employ additional methods, but it would definitely strengthen the paper.
Advancements:
This is a useful catalogue of potential DNA binding proteins of interest, beyond just CTCF. Some known TFs are there, but also new ones are found.
Audience:
Basic research mainly, with particular focus on chromatin conformation and TF binding fields.
My expertise:
ML/AI methods in genomics, TF binding models, epigenetics and 3D chromatin interactions.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this work, the authors describe a deep learning computational tool to identity binding motifs of DNA binding proteins associated to insulators that led to the discovery of 99 motifs related to insulation. This is in turn related to chromatin architecture and highlight the importance of directional bias in order to form chromatin loops.
In general, there are some aspects to be clarified and better explored to make stronger conclusions. In particular, there are some aspects to clarify in the text about the Machine Learning procedure (see my points below). In addition, I have some general questions about the biological implications of the discussed findings, listed in detail in the following list.
Also, I encourage the authors to integrate the current presentation of the data with other (published) data about chromatin architecture, to make more robust the claims and go deeper into the biological implications of the current work. Se my list below.
It follows a specific list of relevant points to be addressed:
Specific points:
In this work, the authors describe a deep learning computational tool to identity binding motifs of DNA binding proteins associated to insulators that led to the discovery of 99 motifs related to insulation. This is in turn related to chromatin architecture and highlight the importance of directional bias in order to form chromatin loops.
In general, chromatin organization is an important topic in the context of a constantly expanding research field. Therefore, the work is timely and could be useful for the community. The paper appears overall well written and the figures look clear and of good quality. Nevertheless, there are some aspects to be clarified and better explored to make stronger conclusions. In particular, there are some aspects to clarify in the text about the Machine Learning procedure (see list of specific points). In addition, I have some general questions about the biological implications of the discussed findings, listed in detail in the above reported points.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The study by Osato and Hamada aims at computationally identifying a set of novel putative insulator-associated DNA binding proteins (DBPs) via estimation of their contribution to the expression of genes in the same chromosome region of their binding sites (+- 1Mbp from TSS). To achieve this, the authors leverage a deep learning architecture already published via which ChIP-seq peaks of DBPs in the TSS of a given gene are used to predict its expression level in four human cell lines.
Building on this, the authors used another tool called DeepLIFT to evaluate the weight of each DBP binding site on the final gene expression value. Hence they made the assumption that if a given DBP had an insulator function they could restrict the prediction of the gene's expression to the region included between pairs of that DBP binding sites, and evaluate the pair's motif directionality bias in the distribution of weights. They exemplify their approach's validity by the fact that they can predict the known directionality bias of CTCF/cohesin-bound sites as the highest of the lot, with the F-R orientation of the pairs the most enriched, recapitulating what already known in literature: i.e., that F-R chromatin interaction peaks are the most enriched. In addition, they find several new DBPs showing significant directionality bias; hence they could be candidates for insulation activity. They then provide correlation between these putative insulator binding sites and sites of transition between euchromatin and heterochromatin by independently using histone mark and gene expression datasets. This, of course, is not surprising because (a) there is insulation between regions with heterotypic chromatin identities, and (b) it was already known from the first papers describing insulated chromatin domains that their boundaries were well-enriched for active transcription and transcriptional regulators (e.g., Dixon et al, Nature 2012).
Finally, they use chromatin interaction (looping) sites to check the overlap between CTCF and all other DBPs and define a subset of putative insulator DBPs not overlapping CTCF peaks, suggesting potentially new insulatory mechanisms. These factors were all known transcriptional activators, but this part of the findings carry most of the novelty in the work and have the potential of opening up new directions for research in chromatin organization.
Overall, the methodology applied here is adequate, clear, and reproducible. The major issue, in our view, is that the entire manuscript's findings relies on the usage of deepLIFT, a tool which was not benchmarked previously or by the current study. In fact, deepLIFT is public as regards its code, and also appears as a preprint from 2017 on biorXiv and published in the Proceedings of Machine Learning Research conference. Also, this key tool was developed by the Kundaje lab (who produce high quality alogrithms), and not by the authors. Therefore, the manuscript is predominantly based on the execution of existing workflows to publicly-available data. This does not take anything away from the interesting question posed here, but at the same time does not provide the community with any new algorithm/workflow.
Finally, although I appreciate that the authors are purely computational and have likely no capacity for experimental validation of their claims of new DBPs having insulator roles, I would assume that there are RNA-seq and/or ChIP-seq data out there produced after knockdown of one or more of these DBPs that show directional positioning. Using this kind of data, effects on gene expression can at least be tested in regard to the authors' predictions. Moreover, in terms of validation, Figure 6 should be expanded to incorporate analysis of DBPs not overlapping CTCF/cohesin in chromatin interaction data that is important and potentially more interesting than the simple DBPs enrichment reported in the present form of the figure. Critically, I would like to see use of Micro-C/Hi-C data and ChIP-seq from these factors, where insulation scores around their directionally-bound sites show some sort of an effect like that presumed by the authors - and many such datasets are publicly-available and can be put to good use here.
As secondary issues, we would point out that:
The scientific novelty of the work lies primarily in the identification of a set of DBPs that are proposed to confer insulator activity genome-wide. This has been long sought after in human data (whilst it is well understood and defined in Drosophila). The authors produce a quantitative ranking of the putative insulation effect of these DBPs and, most importantly, go on to identify a smaller subset that are apparently non-overlapping with anchors of CTCF-cohesin loop anchors; the presence of strong motif orientation biases in many DBPs can also be of broad interest, especially those that cannot be trivially ascribable to the loop extrusion process.
However, although these findings open the way for speculation on multiple insulation mechanisms via proteins with multiple regulatory functions, the manuscript provide no experimental or computational means to test the proposed roles of these DBPs - and, as such, this limits the potential impact of the work and mostly targets researchers in the field of genome organization that can test these findings. Having said this, if validated, this work can significantly broaden our understanding of how chromatin is organized in 3D nuclear space.
I typically identify myself to the authors: A. Papantonis, expertise in 3D genome architecture, chromatin biology, and genomics/bioinformatics.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
The authors provide a detailed characterization of the tumor microenvironment (TME) of 91 ovarian cancer patients, broken down in long and short-term survivors (post 5 years). The focus on the role of a subgroup of T cells, gamma/delta γδ) T cells with reported anti but also pro tumorigenic properties, Prior work of the lab has established a link between a subgroup of γδ T cells expressing CD73 and poor prognosis, due to the ability of these cells to produce immunosuppressive cytokines, such as IL10 or IL8 and the production of adenosine, by CD73, in the micromilieu. The data is further backed up by the analysis of fresh tumor specimens and tissue culture work.
Here they continue this story by investigating the TME using tumor microarrays (91 samples), single cell RNA seq (12 patients), imaging mass cytometry (> 30 samples) and flow cytometry (form confirmatory purposes) to define cellular neighborhoods of CD73+ and CD73- γδ T cells. This revealed differences in cellular composition and spatial transcriptome analysis further helped to define the transcriptomes in γδ T cells, cancer cells and cancer associated fibroblasts.
The authors conclude the in ovarian cancer γδ T cells expressing CD73 dampen anti-tumor immunity and propose detection and evaluation of CD73+ γδ T cells as prognostic marker.
The manuscript is well written, and despite its descriptive nature, easy to follow. Data is presented in a clear and easy to read fashion.
Reviewer #1 (Significance (Required)):
Using a well characterized cohort of ovarian cancer patients with detailed clinical follow up the authors report on the predictive power of a subset of γδ T cells expressing CD73, with immune suppressive / regulatory capacity, reading out patient survival in high grade serous ovarian cancer, a still deadly disease. As such the identification of reliable markers predicting survival is a clear medical need. These findings contrast others made in different solid cancers, suggesting tumor type specific differences, which are only starting to emerge, but are of clear clinical relevance.
What is unclear to me and needs to be addressed, is if these patient specimens were taken before or after initial therapy, whether the samples have been stratified according the treatment that they got, assuming it will be mostly platinum compounds (but maybe not), and that the p53 status of the tumors are (if genetics are available this would help to add some granularity to the study that, as it stands is largely descriptive, even though with extremely high resolution. This data should be available and could be integrated.
We thank the reviewer for this insightful and constructive comment. We agree that clinical context and treatment stratification are essential to strengthen the interpretation and translational value of our findings.
We confirm that all tumor samples used in this study were obtained prior to any systemic treatment, i.e., before first-line chemotherapy, during the Biopsy realized for the diagnosis. This information has now been clearly stated in the Methods and Results section (page 4, line 103) and also in Table S1.
Although our primary aim was not to evaluate correlations with mutational status, we recognize the critical role that tumor genetics play in shaping the immune microenvironment. Using available clinical genomics data, we found that the TP53 mutational status of our cohort aligns with that of previous analyses. As expected for high-grade serous ovarian cancer (HGSOC), nearly all tumors exhibited TP53 mutations (present in 95% of patients). Due to the lack of variability in TP53 status, no meaningful stratification was observed based on this factor. This information has been added in the Materials and methods part (page 4 lines 104 to 106)
Some minor issues
We appreciate this suggestion. To clarify the nomenclature and avoid confusion, we have consistently indicated throughout the text and figure legends that NT5E refers to the CD73 gene.
In the manuscript, the term “density” specifically refers to the density of γδ____ T cells and not the density of CD73 molecules expressed by these cells. Additionally, it is not feasible to conduct a density analysis of molecules using the data obtained from immunofluorescence (IF) staining of sample sections.
Kaplan-Meier analyses were performed to assess patient survival based on the density of total γδ____ T cells, as well as the subsets of CD73⁺ and CD73⁻ γδ T cells. The results indicate that a higher density of γδ____ T cells is associated with poorer patient survival, with a more pronounced effect seen in those with a high density of CD73⁺ γδ T cells compared to those with CD73⁻ γδ T cells.
As the reviewer pointed out, patients with a low density of CD73⁺ γδ T cells do not show significantly different survival outcomes compared to those with a low density of CD73⁻ γδ T cells (IC50 for low CD73⁺ = 6.0 years vs. IC50 for low CD73⁻ = 6.2 years). In response, we have revised the corresponding sentence in the text and included the IC50 values for greater clarity and informativeness (page 9).
The modification has been done.
We appreciate the feedback on figure presentation. We have now updated Figure 2 with improved labeling, consistent font size, and enhanced resolution to ensure better readability across all panels, particularly panels B–D. The revised figure has been updated in the main manuscript.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
In this manuscript ("Deciphering the tumor-infiltrating CD73+ regulatory γδ T cell ecosystem associated with poor survival of patients with ovarian cancer"), Chabab et al. report on the phenotype and location of CD73+ γδ T cells in ovarian cancer. CD73+ γδ T cells can be immunosuppressive via the production of cytokines (IL-8, IL-10) and the expression of PD-L1. Here, the authors investigated the phenotype and location of CD73+ and -neg γδ T cells in ovarian cancers with a particular focus on the cells surrounding the γδ T cells in the tumour.
Overall, the study is informative and well-performed. However, the way some of the data are presented does not allow to fully evaluate them. Besides this, this reviewer only has some minor comments.
General comments:
We thank the reviewer for this thoughtful point. We have amended the text to make it consistent with the data.
As requested by the reviewer, we have revised the figure legends to make them more explicit. We have indicated the number of biological replicates (n) and how many times each experiment was performed independently. This information has been added to each legend where consistent and relevant, to ensure clarity and reproducibility.
This point has been addressed as requested by the reviewer.
All these points have been addressed.
M&M - Please be consistent, if you provide catalogue numbers or dilutions (antibody, reagents) [which is good, maybe even adding the RRID number], do so for all items.
This point has been addressed as requested by the reviewer.
This point has been added in M&M section.
We have amended the protocol of IL-6 Elisa in M&M section for clarification.
Figures Fig.1: - The authors used the word 'predict' in the heading, which seems not appropriate for a retrospective study; something like 'correlate' seems better.
The word “predict” has been replaced by “correlate” as suggested by the reviewer.
The title of the figure has been modified
Pictures of IF have been added as Supplementary Fig 1.
The figure has been corrected.
Fig.2 - It seems funny to call the patients 'naïve', maybe 'untreated' is clearer.
We appreciate this suggestion and agree that ‘untreated’ is a clearer and more appropriate term in this context. We have replaced all instances of ‘naïve’ with ‘untreated’ throughout the manuscript to avoid ambiguity.
We thank the reviewer for this valuable observation. In response, we have replaced the original visualization in Figure 2E with grouped bar graphs showing the mean ± SEM of the relative proportions of each major cell type in the NT5E_low and NT5E_high groups, based on the median split. This format allows for clearer visual comparison of cell frequencies across conditions.
Furthermore, we performed statistical comparisons using a t-test (a parametric test) on each population to evaluate differences in cell type proportions between the two groups. The results indicate a significantly higher proportion of CAFs and γδ T cells in the NT5E_high tumor profile. The corresponding p-values are provided in the figure legend. We hope this revised analysis and clearer presentation address the reviewer’s concerns.
Fig.3 - For Fig3b+c, the IMC are derived from 4 patients (not clear for the flow data)
As stated in both the figure legend and the text, the IMC analysis was conducted on 38 ROIs from four patient samples, while the flow cytometry analysis was performed on tumor samples from seven ovarian cancer patients.
"As shown in new Figures 3b and 3c, no significant differences were observed between patients. Each individual patient is represented by a different color."
The text describing Fig. 3e has been amended in the new version of the manuscript.
The protocol has been amended in the “Materials and Methods” section. A gating strategy and primary data analysis from one representative patient are included in a supplementary Figure 4c.
We agree with the reviewer’s comments that it is surprising that γδ T cell stimulation is not required for IL-8, IL10 and IFNγ production. However, one possible explanation is the high reactivity of γδ T cells compared to other T cell subsets, as well as their localization in the tumor microenvironment rather than in healthy tissue or blood.
This analysis shows the percentage of cells that are positive for both IL-8 and IL-10.
The figure and its legend have been amended for clarity.
Fig.4 - Please provide the values and the statistical analyses for all cell populations.
We performed statistical analyses (Wilcoxon signed-rank test) for all cell populations and provide the data in the Supplementary Fig. 5A. However, due to the heterogeneity of ROIs, a significant difference was observed for tumor cells, which were more prevalent more in the neighborhood of CD73- than CD73+ γδ T cells (p
Fig.5/6 - In Fig5, the authors state that 8 cell populations were differentially enriched around CD73+ or -neg γδ T cells. However, in Fig4, only 4 of these populations are mentioned. Please add the remaining 4 to fig4 and name the 8 clusters in fig5 in line with the gating strategy used in fig4.
We thank the reviewer for highlighting that the description of Figure 5 in our text was unclear. We have revised the text for clarification and specify that based on Supplementary Figure 7, which shows the number of cells for each cell type found in the neighborhood of all γδ T cell subsets (CD73- and CD73+) in all ROIs. We decided to perform phenotypic analysis on only four cell types (those with a sufficient cell counts), setting the cutoff at 700 cells.
The four cell types are analyzed in Figures 5 and 6. Figure 5A shows tumor cells, with eight clusters identified, while Figure 5B represents fibroblasts, with seven clusters identified. Figure 6A shows CD4 T cells, with eight clusters, and Figure 6B CD8 T cells, with ten clusters.
We have added the code color of tSNE plots in Figures 5, 6, and SF9. The tables in Supplementary Figure 8 show the percentage of cells in each cluster within the vicinity of CD73+/- γδ T cells, allowing for an investigation of the neighborhood of each γδ T cell subset.
As requested by the reviewer, we have amended the text to clarify that: “Cluster analysis revealed that CD4+ T cells in contact with effector γδ T cells (i.e., the CD73- subset) express HLA-DR and/or PD-1, both activation markers.”
Supplements - SF2a: please check the labels; how can CD8+ CD4+ cells be labelled 'CD8 T cells' and why do the authors exclude the possibility that e.g. B cells could express HLA-DR?
We thank the reviewer for pointing out the error in Figure 2a, which has now been corrected. The CD8+ cells have been relabeled as 'CD8 T cells,' and the B cells are now shown expressing HLA-DR.
We believe the reviewer is referring to SF9 rather than SF7 in this comment. SF9 analyzes γδ T cells in proximity to CD73+ and CD73- γδ T cells. As in Figures 5 and 6, γδ T cell neighbors of CD73+ and CD73- γδ T cells were identified, and a clustering analysis revealed five distinct clusters. Tumor cells was not analyzed in this figure. We have clarified the text to prevent confusion
As requested by the reviewer, we have performed statistical analysis for SF9b and added a negative control. Additionally, we have included summary data with a statistical analysis in SF9c.
We have updated Supplementary Figure 9B to provide more robust data. We thank the reviewer for pointing out our error. The publication we intend to cite is a research article, not a review.” Hu G, Cheng P, Pan J, Wang S, Ding Q, Jiang Z, et al. An IL6-Adenosine Positive Feedback Loop between CD73+ γδ Tregs and CAFs Promotes Tumor Progression in Human Breast Cancer. Cancer Immunol Res. 2020;8:1273–86.” we made the correction in the manuscript.
Reviewer #2 (Significance (Required)):
In this manuscript ("Deciphering the tumor-infiltrating CD73+ regulatory γδ T cell ecosystem associated with poor survival of patients with ovarian cancer"), Chabab et al. report on the phenotype and location of CD73+ γδ T cells in ovarian cancer.
CD73+ γδ T cells can be immunosuppressive via the production of cytokines (IL-8, IL-10) and the expression of PD-L1. Here, the authors investigated the phenotype and location of CD73+ and -neg γδ T cells in ovarian cancers with a particular focus on the cells surrounding the γδ T cells in the tumour.
Overall, the study is informative and well-performed. However, the way some of the data are presented does not allow to fully evaluate them. Besides this, this reviewer only has some minor comments.
To enable a full evaluation of the data, we have added new figures, amended others, and clarified certain points in the text, hoping that the reviewer will find these modifications sufficient to consider our manuscript for publication.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
In this article, Chabab et al. analyze sample from ovarian cancer patients, with a specific focus on gamma-delta T cells (Tγδ). The authors claim that CD73+ cells are associated with poor prognosis in ovarian cancer, and that CD73 expression is correlated with the composition and polarization of the microenvironment. Using imaging mass cytometry data, they also claim that the neighborhoods of CD73+ and CD73- Tγδ cells differs in composition.
Major comments: - The prognostic value of CD73/NT5E is analyzed in TCGA-Ovarian RNAseq data. In the context of this article, it is implied that this should reflect CD73 expression by Tγδ but it is likely that other cell types are contributing to bulk CD73 expression.
We appreciate the reviewer’s insightful comment. In fact, due to low proportion of Tγδ in TME we have stratified on NT5E total expression. We agree that this signal likely includes contributions from multiple cell types beyond γδ T cells, such as cancer-associated fibroblasts and endothelial cells, which are also known to express CD73 (NT5E gene).
The stratification of patient based on NT5E total expression showed an association between high NT5E expression and poorer overall survival and increase in Tγδ gene markers (TRDC, TRGC1/2) and percentage of cells (Fig2E) in the patient cohort (Fig2C). To clarify this point, we have revised the Results and Discussion sections to explicitly state that the TCGA-based survival analysis reflects total intratumoral NT5E enrichment and cannot be attributed specifically to γδ T cells. We now refer to this analysis as an independent validation of the clinical relevance of CD73, while noting that its cell-type-specific contribution remains to be resolved in future studies using spatial transcriptomics or deconvolution approaches.
We thank the reviewer for raising this critical point regarding potential batch effects and dataset-driven bias in our stratification strategy. To address this, we performed additional analyses to assess whether NT5E (CD73) expression is confounded by dataset of origin.
First, we verified that all single-cell datasets (GSE147082, GSE241221, and GSE235931) were processed using a harmonized integration workflow, including SCTransform normalization and integration using Seurat’s reciprocal PCA approach, which effectively minimizes batch-related variability.
We appreciate the reviewer’s insightful comment. We have added data and updated Supplementary Figure 9 to provide more robust findings. Regarding the role of IL-6, our data in ovarian cancer are consistent with the study by Hu et al. in breast cancer, which reports an IL-6-Adenosine Positive Feedback Loop between CD73+ γδ Tregs and CAFs that promotes tumor progression in human breast cancer."
Minor comments:
The threshold has been added in figure and text.
The correction of figure has been made.
These points have all been amended and corrected in the next version of the manuscript.
Reviewer #3 (Significance (Required)):
This paper shows interesting imaging mass cytometry data of ovarian cancer specimens. The focus on CD73 expression by Tγδ is fairly specific, although the exonucleotidases pathway involving CD73 is currently extensively studied for its immunosuppressive role.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this article, Chabab et al. analyze sample from ovarian cancer patients, with a specific focus on gamma-delta T cells (Tgd). The authors claim that CD73+ cells are associated with poor prognosis in ovarian cancer, and that CD73 expression is correlated with the composition and polarization of the microenvironment. Using imaging mass cytometry data, they also claim that the neighborhoods of CD73+ and CD73- Tgd cells differs in composition.
Major comments:
Minor comments:
This paper shows interesting imaging mass cytometry data of ovarian cancer specimens. The focus on CD73 expression by Tgd is fairly specific, although the exonucleotidases pathway involving CD73 is currently extensively studied for its immunosuppressive role.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this manuscript ("Deciphering the tumor-infiltrating CD73+ regulatory γδ T cell ecosystem associated with poor survival of patients with ovarian cancer"), Chabab et al. report on the phenotype and location of CD73+ gd T cells in ovarian cancer. CD73+ gd T cells can be immunosuppressive via the production of cytokines (IL-8, IL-10) and the expression of PD-L1. Here, the authors investigated the phenotype and location of CD73+ and -neg gd T cells in ovarian cancers with a particular focus on the cells surrounding the gd T cells in the tumour. Overall, the study is informative and well-performed. However, the way some of the data are presented does not allow to fully evaluate them. Besides this, this reviewer only has some minor comments.
General comments:
M&M
Figures
Fig.1:
Fig.2
Fig.3
Fig.4
Fig.5/6
Supplements
In this manuscript ("Deciphering the tumor-infiltrating CD73+ regulatory γδ T cell ecosystem associated with poor survival of patients with ovarian cancer"), Chabab et al. report on the phenotype and location of CD73+ gd T cells in ovarian cancer. CD73+ gd T cells can be immunosuppressive via the production of cytokines (IL-8, IL-10) and the expression of PD-L1. Here, the authors investigated the phenotype and location of CD73+ and -neg gd T cells in ovarian cancers with a particular focus on the cells surrounding the gd T cells in the tumour. Overall, the study is informative and well-performed. However, the way some of the data are presented does not allow to fully evaluate them. Besides this, this reviewer only has some minor comments.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The authors provide a detailed characterizsation of the tumor microenvironment (TME) of 91 ovarian cancer patients, brokend down in long and short term survivors (post 5 years). The focus on the role of a subgroup of T cells, gamma/delta (gd) T cells with reported anti but also pro tumorigenic properties, Prior work of the lab has established a link between a subgroup of gd T cells expressing CD73 and poor prognosis, due to the ability of these cells to produce immunosuppressive cytokines, such as IL10 or IL8 and the production of adenosine, by CD73, in the micromilieu. The data is further backed up by the analysis of fresh tumor specimens and tissue culture work.
Here they continue this story by investigating the TME using tumor microarrays (91 samples), single cell RNA seq (12 patients), imaging mass cytometry (> 30 samples) and flow cytometry (form confirmatory purposes) to define cellular neighborhoods of CD73+ and CD73- gd T cells. THis revealed differences in cellular composition and spatial transcriptome analysis further helped to define the ttranscriptomes in gd T cells, cancer cells and cancer associated fibroblasts.
The authors conclude the in ovarian cancer gd T cells expressing CD73 dampen anti-tumor immun ity and propose detection and evaluation of CD73+ gd T cells as prognostic marker.
The manuscript is well written, and despite its descriptive nature, easy to follow. Data is presented in a clear and easy to read fashion.
Using a well characterized cohort of ovarian cancer patients with detailed clinical follow up the authors report on the predictive power of a subset of gd T cells expressing CD73, with immune suppressive / regulatory capacity, reading out patient survival in high grade serous ovarian cancer, a still deadly disease. As such the identificaiton of reliable markers predicting survival is a clear medical need. These findings contrast others made in different solid cancers, suggesting tumor typ specific differences, which are only starting to emerge, but are of clear clinical relevance.
What is unclear to me and needs to be addressed, is if these patient specimens were taken before or after initial therapy, whether the samples have been stratified according the treatment that they got, assuming it will be mostly platinum compounds (but maybe not), and that the p53 status of the tumors are (if genetics are available this would help to add some granularity to the study that, as as it stands is largely descriptive, even though with extremely high resolution. This data should be available and could be integrated.
Some minor issues
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
The authors do not wish to provide a response at this time
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The authors describe a modified version of single molecule Fluorescence In-situ Hybridization (smFISH) method they have adapted to successfully measure RNA levels in isolated human donor T cells, that are very hard to grow on glass and have small amounts of cytoplasm relative to cell size, a challenge for all researchers working with small cells that only grow in suspension cultures. Using this methodology, the authors have queried transcription status and mRNA localization and fate of the two cytokines, IFNG and TNF, upon T-cell activation. The main findings of the study are: (1) activation of T-cells results in rapid accumulation of IFNG an TNF mRNA; there is differential distribution of the cytokine mRNAs between the nucleus and cytoplasm with greater accumulation in the cytoplasm as activation progresses resulting in increased protein production. There is significant transcriptional heterogeneity in response to T-cell activation. (2) The cytokine mRNA turnover appears to be controlled by translation. (3) HUR, an RBP appears to control poly(A) tail length of TNF mRNA in response to T-cell activation. The successful implementation of a modified smFISH protocol used in this study is a welcome resource for all labs that want to study small human primary cells that are difficult to culture on glass coverslips and grow as suspension cultures. Although the authors have very exciting observations, they have shied away from discussing their results in the context of the biology of T-cell activation and how their observations may explain prior studies on cytokine gene expression patterns during T-cell activation.
In my opinion, the authors should discuss their observations in depth from the context of T-cell activation and cytokine expression. I have enumerated several specific comments that may help the authors in revising the manuscript if they choose to do so.
Specific comments:
Minor comment:
Referee's cross-commenting
I must confess I am not an immunologist, so my knowledge of the intricacies of gene expression in T-cells in very limited. However, I do have a fair sense of transcription regulation and use single molecule approaches, especially smFISH, to address these questions. I agree with the other reviewer the study is of significance, especially the advancement in the ability to do smFISH in primary cells, a challenge that I know first hand. I also have to agree with the other reviewer that the discussion was too short and the authors shied away from the bigger picture of being able to comment on regulation of expression of cytokines during T-cell activation. It is remarkable that they see heterogeneity in gene expression of the individual target genes and bi-allelic expression. The other point of interest is the difference in p(A) tail length and its potential role in regulating TNF gene expression.
The successful implementation of a modified smFISH protocol used in this study is a welcome resource for all labs that want to study small human primary cells that are difficult to culture on glass coverslips and grow as suspension cultures.
Overall, this work is of high quality and can be better presented to fully explore and discuss the biological implications of the observations from the study. It is not clear to me if the authors wished to present this manuscript reporting an advancement in technology tool to study gene expression during T-cell activation, or a more in-depth study of gene expression.
The study will benefit the larger community that use single molecule approaches to understand genew expression.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript by Lattanzio and colleagues uses advanced single molecule FISH, adapted specifically for T cells, to examine RNA transcriptional dynamics in polyclonally stimulated human T cells. By examining the subcellular localisation of both IFNg and TNF mRNAs (nascent and mature), they are able to characterise rate things like rate of transcription and RNA stability. Key findings include the identification of bi-allelic vs mono-allelic transcription at the single cell level which maps to polyfucntion vs monofunctional T cells. Moreover, they identified distinct mechanisms regulating RNA stability and the role of RNA binding protein HuR in mediating that.
Overall, this is really a proof of concept paper that uses elegant technologies and analysis tools showing just how much information can be obtained from this approach. The ability to examine RNA dynamics and the imapct of RNA binding proteins in regulating RNA stability/translation/transriptoin at a single cell level will be an advance for the field, not just those interested in T cell biology but all cell types.
There are no specific experimental issues that came to mind that need to be addressed and it is really only some minor comments, particularly for the discussion that would strengthen the implicaitons of the study.
A minor point relates to line 329, that sentence stating "Even though most activated Teff cells express cytokine mRNAs, they display a two order of magnitude difference in mRNA and protein expression."
It is not clear what this is relevant or compared to. A two order of magnitude difference compared to what?
This is a proof of concept study that demonstrates the utility of the T cell smFISH approach to delineate high resolution analysis of cytokine RNA dynamics at a single cell level, for multiple cytokine RNA species. It clearly provides interesting biology and further understanding of RNA dynamics in activated T cells. I especially appreciated the observation of bi-allelic vs mono-allelic transcription, and the ability to explore the role of RNA binding proteins in RNA regulation.
This technique will have broader applicability and hence will be of interest to those outside T cell immunology. It only requires some minor corrections/revisions.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer #1 (Significance (Required)):
This study aims to bridge a gap between the mechanisms of preeclampsia and neurodegenerative disorders, and this through the existence of misfolded proteins in the preeclamptic placenta which has been reported before, in particular the beta amyloid protein, known as operative in Alzheimer's disease (AD) in particular.
Our response: We sincerely appreciated the reviewer’s constructive comments.
*Reviewer #1 (Evidence, reproducibility and clarity (Required)): *
Minor remarks
It is classical now to present in extenso the WB as supplementary data for Fig 3, 4 and 5. Our response: We will include the full blots in Supplemental information.
*
It seems that the beta amyloid signal is not stronger for the early onset and the late onset PE samples. Have the authors an interpretation?
Our response: The current manuscript includes both early-onset and late-onset cases. Thus, we are certain that amyloid beta deposition is involved in both early- and late-onset PE. We will discuss this matter.
The figure 4b does not show the BeWo labeling in forskolin with or without beta amyloid peptides, why? It would be illustrative to show a decrease in the fusion processes
Our response: In Fig. 4A, we pretreated BeWo cells with Aβ fibrils and after that, cell fusion was induced by Fsk. On the other hand, in Fig. 4b, we treated BeWo with Aβ fibrils and investigated the protein levels and subcellular localization of ZO-1 and E-cadherin. Fig.4b shows that expressions of proteins involved in cell-cell interaction were reduced by Aβ fibril treatment without Fsk. Cell-cell interaction before syncytialization is required for cell fusion, and these proteins disappear after cell fusion. Thus, our results demonstrate that elimination of cell-cell interaction by Aβ fibrils resulted in reduced cell fusion induced by Fsk. This is why we treated BeWo cells with Aβ fibrils before the induction of cell fusion by Fsk, and BeWo labeling in forskolin with or without Aβ fibrils will result in a loss of ZO-1 and E-cadherin regardless of the occurrence of cell fusion. We will discuss this matter in more detail.
How do the authors explain that exposure to fibrils did not seem to slow down significantly the fusion process, even though markers are decreased?
Our response: Since we previously demonstrated that loss of membrane E-cadherin slows the fusion (Iwahashi et al., Endocrinology, 2019, PMID: 30551188), we believe that reduction of membrane localization of E-cadherin also slows the fusion process. We will discuss this matter further.
Could the authors attempt a labeling with the Di-8, an interesting quantitative marker of cell fusion (see ref PMID: 38019394).*
*
Our response: We have shown that pretreatment of BeWo cells and human primary cytotrophoblasts (CTBs) inhibited induction of syncytin-1 and β-hCG. Syncytin-1 is a critical driver of syncytialization and formation of the syncytiotrophoblast layer, and β-hCG is one of the major products of syncytiotrophoblasts. Thus, induction of these proteins is widely used as syncytialization markers of trophoblasts. On the other hand, Di-8-ANEPPS is a potentiometric fluorescent dye that may be used assess cell fusion simply and economically. Although we understand the robustness of this method, we believe that the current data are sufficient to demonstrate that Aβ fibril pretreatment inhibited syncytialization of BeWo cells and CTBs.
* *
Reviewer #2 (Significance (Required)):
Investigating the deposition of Aβ in the placenta could enhance our understanding of pregnancy complications such as PE, fetal growth restriction, and neurodevelopmental risks. However, further research on this topic is necessary.
Our response: We sincerely appreciate the critical reading and constructive comments of the reviewer. We agree that further research on protein aggregation and the pathogenesis of preeclampsia is necessary. We will discuss this matter in the discussion.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
Major comments
Our response: We thank the reviewer for the critical comment. We investigated Aβ generation by an EVT model cell, HTR8/SVneo cells. We found that HTR8/SVneo cells produced much less amount of Aβ compared to BeWo cells (unpublished). Gao et al. reported that Aβ aggregates induced autophagy in HTR8/SVneo cells and suggested that an excessive autophagy may be detrimental and be involved in the development of preeclampsia (Gao et al., J Mol Histol, 2024, PMID: 38777993). We will discuss this matter in the discussion.
On the other hand, we have already investigated the effects of Aβ monomers in EVTs, and discovered that even low levels of Aβ produced by EVTs promote EVT invasiveness and have a physiological function. Please see below. We will add these new data in the revised manuscript.
*
*
Our response: We did not observe any pathologies near the Aβ deposition.
Our response: We agree with the reviewer. There is a time discrepancy between HIF activation and BACE1 induction. Our immunohistochemical analysis showed that PE placentas are in a chronic hypoxia condition and that BACE1 was increased in PE placentas. Our cell-based assay supports that HIF1α stabilization by Roxadustat increased BACE1 levels in BeWo cells. We will tone down the results section of the immunohistochemical analysis.
*
Our response: Roxadustat inhibits the proline hydroxylation of HIF1α and thereby inhibits the ubiquitination and degradation of HIF1α via the ubiquitin proteasomal system. In this study, we used Roxadustat as a HIF1α stabilizer to investigate whether BACE1 levels are increased with hypoxia and HIF1. Our data showed that treatment of BeWo cells with Roxadustat increased HIF1α levels, supporting the efficacy of Roxadustat. We will include this information in the result section for clarity.
Our response: To date, no report has been found showing Aβ deposition in placentas other than PE. The deposition of protein aggregates, including those of Aβ and transthyretin, has previously been reported in PE. However, the presence and role of these protein deposits in placentas under pathological conditions, in addition to PE, remains to be elucidated. Several stresses such as hypoxia and ER stress may lead to deposition of protein aggregates in the placenta. These points will be discussed in the discussion.
*
Our response: We did not find any other complications in the normal placentas. In the brain, Aβ is constitutively generated and thus, thought to play physiological roles. The amount of Aβ is determined by the balance between the production and the clearance. A sustained imbalance of Aβ production and Aβ clearance will lead Aβ aggregation and deposition. We found that BeWo cells expressed BACE1 in a normoxic condition and thus, normal placentas may express BACE1 and generate small amounts of Aβ. Our results suggested that chronic hypoxia in PE placentas resulted in increased BACE1 expression and increased Aβ production, which may eventually result in Aβ aggregation and deposition, because the aggregation process of Aβ is concentration-dependent. We will include this point in the revised manuscript.
*
Our response: We used RB4CD12 as a protein aggregation marker. As shown in Table 1, the current study includes 3 placentas whose gestational ages are over 37 weeks. We did not observe RB4CD12 and Aβ deposition in gestational age-matched control and observed BACE1 expression in one 37 weeks gestational age control. We will include these points in the result section.
*
Our response: At the early gestation, physiological hypoxia promotes the EVT invasion and helps the remodeling of spiral arteries for oxygen supply. Please see our response above. Severe hypoxia on the CTB side in early gestation may result in a miscarriage before PE develops.
Minor comments
Our response: We thank the reviewer for pointing this out. We will correct the methods.
*
Our response: We do not have information about the clone number of BeWo cells used in this study. We purchased them from the American Type Culture Collection (Manassas, VA) and they were authenticated by JCRB Cell Bank (National Institute of Biomedical Innovation Japan, report no. KBN0410). By using the same cells, we published three articles in which we successfully analyzed syncytialization of BeWo cells (Yamamoto et al., Endocrinology, 2017, PMID: 28938427; Iwahashi et al., Endocrinology, 2019, PMID: 30551188; Matsukawa et al., Biomolecules, 2022, PMID: 36008943). We would like to apologize for our mistake in the description of BeWo cells in the methods section and thank the reviewer for providing us with an opportunity to correct our mistake. We will note that BeWo cells were purchased from the American Type Culture Collection (Manassas, VA) and authenticated by JCRB Cell Bank (National Institute of Biomedical Innovation Japan, report no. KBN0410) in the methods section, and will upload the authentication report KBN0410 as a review process file.
*
Our response: We repeated 6 experiments (the repetitions are biological, not technical, replicates). The results are shown as means ± SEM (n = 6) as stated in the Figure legends.
* *
Reviewer #3 (Significance (Required)):
While Aβ is present in human placentas and accumulates in preeclamptic placentas, the production and role of Aβ in the human placenta remain unclear. The current findings suggest that increased Aβ production in cytotrophoblast by hypoxia may lead to the formation of Aβ fibrils, which inhibit syncytiotrophoblast formation and are detrimental to pregnancy, revealing a novel role of Aβ fibrils in the pathogenesis of preeclampsia.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
The authors found that Amyloid β suppressed cytotrophoblasts syncytialization, which is innovative. The authors used human patient samples and human primary CTB culture which are powerful data.
Our response: We appreciate the reviewer’s thoughtful feedback and support.
Fig. 3. The authors used Roxadustat to stimulate HIF-1α and showed BACE1 increase. It would be better to have the cells in real hypoxia condition.
Our response: There is a time discrepancy between the increase in HIF-1α levels by hypoxia and induction of BACE1. Because the purpose of this experiment is to show that increased HIF1-α correlated BACE1 induction, we used Roxadustat as a HIF1-α stabilizer and showed that sustained induction of HIF increased BACE1 levels. However, we do understand the reviewer’s concern. We will include data showing an increase in BACE1 in hypoxic conditions by performing new Western blotting experiments.
Fig. 4 and 5. The authors used external Amyloid β for stimulation. Would the endogenous Amyloid β levels reach the concentration of external one? It would be better to see the quantitative levels of Amyloid β in Fig. 3b.
Our response: Because the aggregation of Aβ requires a high concentration of a micromolar order, we used synthetic Aβ fibrils for stimulation. We propose that chronic hypoxia in preeclampsia leads to an elevated local concentration of Aβ through a sustained increase in Aβ production, which eventually results in Aβ fibrillogenesis and deposition of Aβ fibrils. Therefore, it will be difficult for the Aβ concentrations generated by BeWo cells to reach a level sufficient for fibrillogenesis. We will discuss this point in the revised manuscript. In addition, we have already performed ELISA assays to quantitatively analyze Aβ generation by BeWo cells. We will include these ELISA data in the revised manuscript.
* *
Reviewer #4 (Significance (Required)):
The manuscript addresses an important theme recently identified to address the heterogeneous etiology of preeclampsia. Although the authors have used in vitro approaches, the study could have been a solid if not for some major concerns.
The authors have focused on an already demonstrated phenomenon but have tried to validate the findings using their in vitro approaches. The manuscript is well written but some lapses for correct references.
Our response: We thank the reviewer for the critical reading of our manuscript and his/her constructive comments. As the reviewer pointed out, recent studies suggest that preeclampsia is a proteinopathy. However, the mechanisms by which protein aggregate plays detrimental roles in placentation has not been well-understood. In the present study, we discovered a detrimental role of Aβ fibrils in syncytiotrophoblast formation.
Reviewer #4 (Evidence, reproducibility and clarity (Required)):
Major comments:
Our response: We sincerely appreciate the reviewer for his/her helpful comment. We will revise the introduction by citing the references recommended by the reviewer.
*
Following up on the comments made above, the authors talk about induction of Aβ in hypoxia-treated human trophoblasts represented by an established cell line, BeWo, and primary human trophoblasts. However, it is not clear whether Aβ42 as stated in the manuscript was detected as an aggregated structure or a protein coupled with RB4CD12 aggregate marker. It would have been helpful if the authors could provide direct evidence for Aβ aggregation.*
*
Our response: Based on our previous findings showing that highly sulfated domains of heparan sulfate are common components of protein aggregate deposits, we used RB4CD12, which recognizes these domains, as a marker of protein aggregate deposition. These include aggregates of Aβ in Alzheimer’s disease, transthyretin in ATTR, and p53 aggregates in p53-mutated cancers (Hoshono-fukao et al., Am J Pathol, 2012, PMID: 22429964; Kameyama et al., Am J Pathol, 2019, PMID: 30414409; Iwahashi, PNAS, 2020, PMID: 33318190). Please also see our reply to Comment 5 below. We will perform additional immunohistochemical analysis with the β0001 anti-Aβ antibody and the ProteoStat dye that recognizes protein aggregates.
*
What appeared to be more surprising is the statement on lines 162 and 163 that cultured CTBs produced Aβ40/42. Again, it is not clear whether the authors are talking about aggregated Aβ or just induction of Aβ. Why should normal CTBs produce Aβ? It is not clear whether this is a transient expression or a long-term phenomenon. The issue is distinction between normal and adverse pregnancy conditions, and the latter associated with protein aggregation as suggested in the literature.*
*
Our response: BeWo cells and cultured CTBs produce Aβ peptides in a normoxic condition. In the brain, neurons constitutively produce Aβ peptides, which have physiological roles such as controlling neuronal hyperexcitability, enhancing of synaptic plasticity, and improving memory (reviewed in Kent et al., Acta Neuropathol, 2020, PMID: 32728795). The amount of Aβ in the brain is regulated by the balance between Aβ production and Aβ clearance, and the imbalance of the production and the clearance may result in an increase in Aβ local concentration and Aβ aggregation. Our results showing that hypoxia increased Aβ production in BeWo cells suggest that chronic hypoxia, which is a risk of preeclampsia, may lead to a sustained increase in Aβ production and an elevated local concentration of Aβ at or near the site of Aβ production. We will discuss these points in the discussion.
In the present study, we showed that aggregated form of Aβ (i.e., Aβ fibrils) was detrimental to the CTB differentiation. On the other hand, we already found that Aβ monomers promoted EVT invasion (please see the below). We believe that promotion of EVT invasion by Aβ monomers represent a physiological function of Aβ in the placenta. We will include these new data in the revised manuscript and we will also perform experiments with BeWo cells and Aβ monomers in order to investigate whether Aβ monomers have some roles in CTB differentiation.
*
*
*
The authors have adequately pointed to importance of hypoxia in the onset of preeclampsia-like features. As a matter of fact, Lai et al demonstrated in a mouse pre-clinical model that hypoxia could induce severe features of preeclampsia (Hypertension. 2011;57:505-514). The use of hypoxia as driver of Aβ induction is appreciated.*
*
Our response: We agree with the reviewer that studies using preclinical animal models are an important topic for the future. __We will discuss this point in the discussion. __
*
In Fig. 1, although the authors have used DIC approach, it would have been helpful if they presented individual Aβ and RB4CD12 green and red channels, and a merged profile. For example, PE #4 sample does not appear to have much RB4CD12. Again, there is a question of aggregated or native protein structures. It is difficult to have a satisfactory statistical analysis. Did the authors look for Aβ in the anchoring villi region of the placenta?*
*
Our response: We will show the green and red channel images individually. We have noticed that we detected Aβ deposition without RB4CD12 signals. Aβ is small peptides of 40 to 42 amino acid residues and is extracellularly released after the production. Non-deposited Aβ monomers are not detected by immunohistochemical analysis, because these soluble Aβ peptides are spread out in the tissue fluid. Thus, in our statistical analysis, we calculated only merged signals of Aβ and RB4CD12, which suggests that our data show the aggregated and deposited Aβ. We will note this point in the results. In addition, we will perform immunohistochemical analysis with the anti-Aβ antibody and the ProteoStat dye. Please also see our response to Comment 2 above. We did not observe Aβ deposition at the anchoring villi.
*
Fig. 2 does not show significant staining for HIF1-α in PE placental tissue.*
*
Our response: In a normoxic condition, HIF1-α is constitutively expressed but degraded via the proline-hydroxylation and the subsequent ubiquitination and degradation in the proteasome. Because the proline-hydroxylation is oxygen-dependent, hypoxia induce HIF1-α accumulation. Thus, our data suggest a hypoxic environment in the preeclamptic placentas. We will note this point in the results section.
*
Fig. 3B, why should there be Aβ40/42 under normoxic conditions? This is the most pertinent concern and the authors are validating significant expression of Aβ40/42 under normal conditions. In normal pregnancy placenta, this protein has not been detected.*
*
Our response: Aβ peptides are constitutively produced in BeWo cells, and the production was enhanced by hypoxia. Aβ is small peptides of 40 to 42 amino acid residues. We did not observe Aβ signals in the immunohistochemical analysis of the normal pregnancy placentas, because Aβ peptides that do not aggregate and deposit in the placenta were distributed in the tissue fluid and lost before and during the processing of the placentas for the paraffin-embedding and immunostaining. Our immunohistochemical analysis detects only Aβ deposition. Thus, the absence of Aβ signals in the immunohistochemical analysis of normal placentas does not mean that normal placenta does not produce any Aβ peptides.
*
Figs. 4 and 5 present the crux of the conclusions that the authors are trying to draw from their study. Aβ peptide solution was incubated for 5 days at 370C to prepare so called Aβ fibril-like structures. What is the purity of fibril structures? Does this preparation show toxic effects on cell viability? Human trophoblasts expressing E-cadherin fail to participate in endovascular cross-talk with endothelial cells, a process required for spiral arteries. It appears that either BeWo cells or primary trophoblasts used in this study represent trophoblasts from third trimester. It is not clear why should Aβ fibril like structures should inhibit ZO-1 and E-cadherin or β-hCG (Fig. 5) for that matter. In Fig. 5C, there does not seem to be a major effect of Aβ fibrils. Did the authors try synthetic Aβ as a control. These experiments could have been meaningful but for proper controls.*
*
Our response: Synthetic Aβ was purchased from Peptide Institute (Osak, Japan). The purity is >95%. We will include the data sheet as a review process file. In case that the reviewer wants to know the fibril content of the preparation, we will calculate the fibril content by using Native PAGE followed by Western blotting. We did not observe any cytotoxicity of the preparation as shown in Supplemental Fig. S3.
We previously showed that membrane localization of cell-cell interaction proteins such as ZO-1 and E-cadherin in cytotrophoblasts is required for syncytialization (Iwahashi et al., Endocrinology, 2019, PMID: 30551188; Matsukawa et al., Biomolecules, 2022, PMID: 36008943). Because Aβ aggregates disrupt membrane localization of tight junction proteins partly by inducing excess autophagy (Marco et al., Neurosci Lett, 2006, PMID: 16644119; Chan et al., Exp Cell Res, 2012, PMID: 29856989), we hypothesized that Aβ fibrils may also disrupt membrane localization of ZO-1 and E-cadherin in BeWo cells. We are focusing on the effect of Aβ fibrils on cytotrophoblasts at the late stage of pregnancy when the remodeling of spiral arteries is completed. We understand the importance of investigating the effects of Aβ and Aβ fibrils on early pregnancy. We will cite an article showing the effects of Aβ aggregates on EVTs (Gao et al., J Mol Histol, 2024, PMID: 38777993) and include our data showing the Aβ monomer functions on EVT invasion. Please also see our reply to Comment 3 above. As for Fig. 5C, we will improve the quality of images. We will also perform experiments to investigate whether Aβ monomers alone affect syncytialization of BeWo cells.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript focuses on the role of amyloid β (Aβ) in hypoxia-exposed human trophoblasts. Recent reports in the literature have confirmed the presence Aβ and other proteins, including Tau, transthyretin, and TDP-43, in placental tissue derived from preeclampsia deliveries. These proteins are recognized as hallmark causative factors for Alzheimer's disease related dementias. Hypoxia has also been shown to induce expression of these proteins, including Aβ, in human trophoblasts. In this regard, detection of Aβ hypoxia-exposed human trophoblast may not be a novel finding. This said, the manuscript presents some solid information and could have been very informative study. However, several conceptual, technical and literature concerns remain unaddressed and dampen the reviewer's enthusiasm for this study.
Major comments:
The manuscript addresses an important theme recently identified to address the heterogeneous etiology of preeclampsia. Although the authors have used in vitro approaches, the study could have been a solid if not for some major concerns.
The authors have focused on an already demonstrated phenomenon but have tried to validate the findings using their in vitro approaches. The manuscript is well written but some lapses for correct references.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The authors found that Amyloid β suppressed cytotrophoblasts syncytialization, which is innovative. The authors used human patient samples and human primary CTB culture which are powerful data.
Fig. 3. The authors used Roxadustat to stimulate HIF-1α and showed BACE1 increase. It would be better to have the cells in real hypoxia condition.
Fig. 4 and 5. The authors used external Amyloid β for stimulation. Would the endogenous Amyloid β levels reach the concentration of external one? It would be better to see the quantitative levels of Amyloid β in Fig. 3b.
While Aβ is present in human placentas and accumulates in preeclamptic placentas, the production and role of Aβ in the human placenta remain unclear. The current findings suggest that increased Aβ production in cytotrophoblast by hypoxia may lead to the formation of Aβ fibrils, which inhibit syncytiotrophoblast formation and are detrimental to pregnancy, revealing a novel role of Aβ fibrils in the pathogenesis of preeclampsia.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this manuscript, the authors examine the deposition of amyloid-β (A β) peptides that accumulate in the brains of patients with Alzheimer's disease (AD). The authors demonstrated the expression of HIF-1 in the pre-eclamptic (PE) placental tissue using immunofluorescence (which is not novel), alongside the expression of BACE1. These experiments were also validated using BeWo and primary trophoblast cells cultured under hypoxia to mimic one of the characteristics of PE. However, this manuscript is quite preliminary, and many additional experiments are necessary to confirm the deposition of Aβ fibrils in PE. The authors treated CTB and observed the effects on STB, but in PE, the main cell lineage affected is extravillous trophoblast (EVT) cells, which invade the spiral artery. The defect in this invasion is one of the major causes of PE. Therefore, the authors should investigate the effect of hypoxia and Aβ deposition on EVT invasion. Overall, this work appears very incomplete, and further experiments are warranted.
Major comments
Minor comments
Investigating the deposition of Aβ in the placenta could enhance our understanding of pregnancy complications such as PE, fetal growth restriction, and neurodevelopmental risks. However, further research on this topic is necessary.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Proving that more Beta-amyloid are produced in preeclampsia, and that impacts negatively trophoblast cell fusion is interesting and provides a potential mechanism for interpreting some specific cases of preeclampsia. The authors analyzed the placenta from five control and five preeclamptic pregnancies (4 early onset et 1 late onset).
The authors show first by IHC that amyloid beta and aggregate markers are apparently exclusively detected in the PE samples, the same observation is done for detection of HIF1alpha and BACE1, the enzyme that is responsible for the generation of amyloid peptides from digestion of the APP membrane neuron protein. After having used placental samples, the authors moved to the BeWo cell model, where they could analyze specifically cell biology in the context of syncytialization. The authors inhibited HIF1a prolylation (thus stabilizing it even in normoxia), and this leaded to the increase of BACE1, of beta-amyloid molecules, as shown by WB analyses; the same result was obtained by exposure to hypoxia, while a BACE1 inhibitor had the opposite effect.
An interesting issue is the demonstration provided by the authors that in this model, syncytialization is decreased by Beta-amyloid fibrils, together with decreased hCG expression and decreased Syncytin-1. The authors also validate these results on primary human CTB from the third trimester.
Minor remarks
This study aims to bridge a gap between the mechanisms of preeclampsia and neurodegenerative disorders, and this through the existence of misfolded proteins in the preeclamptic placenta which has been reported before, in particular the beta amyloid protein, known as operative in Alzheimer's disease (AD) in particular.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
*Our lab was totally destroyed on June 15th by an Iranian missile. All stocks, equipment and reagents were lost. While we performed many of the experiments requested by the reviewers, unfortunately some were never completed. We thank you for your understanding. *
We thank the three reviewers for their thoughtful comments and useful suggestions on how to improve our paper. Some of the reviewers claimed that the paper is “preliminary”. We would like to highlight that in our opinion “preliminary” has two possible meanings in this context: 1) the data does not yet support the claims that the authors wrote; 2) the story is short and should be extended. While we totally agree that type 1 “preliminary” should be addressed (and we have addressed that to the best of our abilities), type 2 “preliminary” is a matter of scope, the length of the paper/project and the publication home. We believe that this story, which has been led by an outstanding master’s student (and as such has had a limited timespan) is worthwhile of publication in its current scope.
Reviewers’ comments are in BLUE while our responses are in BLACK.
Reviewer 1 Summary: This study reports a role for matrix metalloproteinases (MMPs) in the developmental pruning of gamma Kenyon cells (KCs) in the fruit fly Mushroom Body during larval-pupal metamorphosis. The authors show through gene expression studies that MMP genes are upregulated in late larval stages as part of the early program for this type of neuronal pruning. They show through cell-targeted RNAi studies of both secreted MMP-1 and membrane-anchored MMP-2, that both genes are required in glial cells and to a lesser extent within KCs.
Both MMPs have secreted and membrane-anchored isoforms and we did not assess whether the secreted/anchored isoforms are involved; e.g. see LaFever et al. 2017.
The authors show that MMP secreted from glial is required for normal levels of Mushroom Body developmental neuronal pruning. They mention that MMP genes have been identified in schizophrenic patient screens in patients, and that perhaps a comparable pruning mechanism could be involved in the loss of grey matter (loss of synapses) in patients. The authors propose that MMP levels may be a potential therapeutic marker in the future.
We thank the reviewer for his comments. We find it important to clarify that we do not think our work suggests that the MMPs levels may be a potential therapeutic marker without much additional work in the future. In the original text we added a claim from another paper suggesting MMPs as therapeutic target. However, due to the arising confusion, we decided to delete this statement from the text (original line 198). We also added a general disclaimer towards the end of the discussion regarding the genetic power of Drosophila but its limited implication into human health (new lines 276-278).
Major Comments: Overall, the work is of a reasonable standard, but very preliminary
Please see general note on two types of “preliminary” – we thank the reviewer for helping us substantiate our claims and strengthen our paper but we do not plan to significantly increase its scope.
The study lacks the substance to completely convince me of any of the results. There is SUBSTANTIAL work that needs to be done to make this publishable. There are a lot of writing mistakes; so many that I do not list them in detail here
We are not absolutely sure that we understand to which mistakes this reviewer is eluding. However, we carefully rewrote the manuscript, streamlined many of our claims and added many new and more recent references.
The references citations are fairly old, but I do not list update replacements here
Thanks – we added many newer and relevant citations.
The text is very brief, and the overall writing needs to include significantly more description and detail
We have included more descriptions and details, as will be elaborated later on, but – again - this is a short report and will remain as such.
This is evident in all aspects of the manuscript, but especially notable in the Methods and Figure Legends
Thanks for raising this comment, which was reverberated also by other reviewers – we have now included more details, with a particular focus on the genotypes (Table 2), that somehow were erroneously not included in the original submission, as well as more detailed figure legends.
None of the Figure Legends include full genotypes of any of the fly lines, and these full fly lines are also not included in the Methods. This is vital to compare the experimental lines to the controls
True – our apologies for this mistake, we now added the full genotypes in Table 2.
Major points are listed below:
- Figure 2: It is important to note of the specific age of animals in these images when talking about the loss of genes in development. Are all the animals age-matched? High levels of synaptic pruning occur post-eclosion), and it is important to understand when these pruning defects occur. It is mentioned that that overlap for the gene expression data is upregulated during 6-18h APF is this when these images are taken? This is very important in the context of pruning as SCZ symptom presentation is very late relative to these early events.
We thank the reviewer for this comment which suggests we were not clear enough in our description. We do not claim to have generated an SCZ model and have clarified this better in the text (lines 275-278). Furthermore, axon pruning happens during pupal development, but in all the main figures in this manuscript we dissected young adult flies (3-5 days post eclosion) and show the remnants of unpruned axons (as we have done in numerous studies). To make sure that initial development occurred normally, we also include larval brains in the Figure S7. We now clarified the fact that we are imaging adult brains as a readout to investigate whether pruning occurred during metamorphosis or not (line 124-126).
- Figure 2: In the figure legend, it is indicated that the arrows are unpruned axons, however in the controls these areas appear to be highly innervated. Further explanation is needed about the context of the arrows, as there are clear visual differences between these images and the controls, but they appear to have a more expansive phenotype than "unpruned axons". The data does not match the visual representation in comparison to the control.
We apologize for this confusion. Unfortunately, the driver which we use to label the γ-axons, R71G10-QF2, is not absolutely specific to the γ type KCs but also expressed (sometimes) in the ɑ/β KCs. As the ɑ/β axons are very stereotypic in shape and also express high levels of FasII (which we stain for), we can easily distinguish between the ɑ lobe and unpruned γ axons. To clarify this point, we now clearly demarcate all lobes in the control images and specifically the ɑ lobe in all panels. Additionally, we added new schemes in Figure 2A and 2O to better clarify the anatomy and experimental design.
- Figure 2: There needs to be more descriptive definitions and clarifications to the defects labeled in panel K. This could be done in the figure legend, but it would be more useful to label the images provided. For example, if Mmp2 is a "mild pruning affect, put that in the pie chart somewhere, to help guide the description of the phenotype to what those confocal images look like.
We understand that the pie chart in Figure 2 was confusing and therefore simplified it in the current version (Fig. 2B and 2P). Also, thanks to this great point, we now include a new Figure S3 that includes examples for the ranking categories, which were now performed by two independent investigators in a blind manner.
Figure 3: The time points of the images of the Mushroom Body (MB) are vital to understanding the process and regulation of these genes.
Please see our comment to point #1 – unless specifically stated otherwise, all images are MBs of adult flies, as now clearly mentioned in the figure legends, in the text and in the Material and Methods section.
- Figure 3D: Significant description of this graph needs to be added for clarity. What parameters separate each phenotypic defect? Labeling the images and showing images that belong in different groups would be very helpful and improve the paper significantly.
We now included a new Figure S3 (also see our response to comment #3).
- Figure S1: Additional experiments would help answer the strength of the phenotype for the ALG-Gal 4 driver. The authors need to perform the rescue experiment. Use a MMP-2 null and then drive it back in the ALG-GAL4 to see if this is sufficient to rescue the neuron pruning. This also isolates the mechanisms to one subtype of glia.
These are excellent suggestions that are, unfortunately, not doable. To perform a rescue experiment, one would need a viable loss-of-function phenotype of an Mmp2 mutant. There is one published Mmp2 loss-of-function null allele which is lethal during pupal development (Page-McCaw et al, 2003). Our previous data, using tissue specific (ts)CRISPR, suggested the involvement of Mmp2 in neurons for their remodeling (Meltzer et al, 2019). We therefore independently generated an Mmp2 germline mutant using CRISPR (harboring an indel resulting in a premature stop codon and predicted to encode a truncated, 77 amino-acid long protein), now described in Fig. S5A (and in the Materials and Methods). This allele is, as expected, unfortunately also lethal. We attempted to overcome lethality by generating MARCM (mosaic) clones in neurons, but as expected, because Mmp2 is largely secreted, there was no pruning defect phenotype (Fig. S5B-C). Unfortunately, it is not yet possible to generate glial clones.
Figure 3 and 4: The other glial subtypes need to be analyze to make any conclusion about their involvement, as well as the involvement of the astrocytes. Running these exact same experiments on the cortex glial and ensheathing glia will provide essential insight into what glial subtype is involved. The presumed lack of phenotypes in these other glial subtypes will also strengthen the argument that the astrocytes are specifically involved in this process. These are vital experiments.
We currently limited our analysis (and conclusions) to astrocytes. Despite the fact that this experiment is beyond our initial scope, we obtained reagents and performed preliminary experiments (using the R77A03-Gal4 driver for cortex glia, and the R83E12-Gal4 for ensheathing glia). In both cases, we observed extremely mild pruning defects, not comparable to those with Repo- or Alrm-Gal4. In these preliminary experiments we lacked a proper control, and now, unfortunately, due to the loss of our lab, we are unable to complete these experiments in a reasonable amount of time.
- Figure 4: Again, description of the phenotypes and examples of these would improve the quality of this figure substantially.
Absolutely agree – see our response to comment #3 (and Fig. S3).
- Figure 5: An improvement on the quantifications of these phenotypes would strengthen the paper substantially. More detailed description of the phenotypes and how they related to the control would significantly improve the overall quality of the work.
Thanks again for highlighting that we neglected to include the full genotypes that are now added (Table 2). We also thank the reviewer for raising the point regarding quantification. First, we generated a new Fig. S3A-E to show examples of the ranking by two independent rankers. Second, ranking was performed by looking at TdTomato positive vertical axons that are outside of the ɑ lobe (high FasII) – this is now better explained in the materials and methods. Additionally, while we would love to have a better scoring, and automatic, system – and even published a semi-automated scoring algorithm in Alyagor et al. 2018 (Figure 3O in the Alyagor paper), because the driver also labels vertical axons (ɑ/β) and because unpruned γ axons often express FasII, this quantification method does not always work. What we have done in previous cases, as we have also done here, is to provide independent ranking by two investigators and compare their ranking (Fig. S3F-G). Finally, we are working with our AI hub to develop automatic scoring systems that will not require human ranking – however this is beyond the scope for this manuscript.
Minor Comments: 1. Figure 1A: I would suggest labeling the KC (gamma) and potentially one of the others (a/B, a'/B') to orient the reader to the differences between these two subsets of the KCs, and to emphasize which neurons are undergoing pruning and where the cell bodies are and where the axons project.
Thanks for the suggestions – we now better annotated the scheme in Figure 1A as well as additional schematics in Figure 2 and, finally, better annotations in selected panels. Specifically, the ɑ lobe is outlined in magenta throughout all relevant panels.
- Figure 1C: This panel needs further labeling to explain the findings in the heat map. Labeling some of the genes that were found and where they were would be helpful. This could also be done in the figure legend, however without any further labeling or context the heatmap is confusing.
We apologize for the incomplete figure. We did not want to overload the figure with data, which is why we are showing only the important clusters and did not include gene names. To keep the figure simple, but at the same time provide the complete information, we now include the full data in Fig. S1 (that includes the original heatmap with all the dynamic clusters I-IX, and including all the gene names). For the full raw data, including non-dynamic clusters, the reader is referred to look in Supplemental excel file 1. We hope this provides the clarity that this reviewer rightfully asks for.
- Figure 3B,C: The full genotypes need to be labeled. What is the exact genotype used for the control?
The full genotypes of all figure panels are now included in Table 2 in the Materials and Methods.
- Figure S1: The stock number for the ALG-GAL4 is missing, there are multiple different drivers, therefore this could be helpful in understanding this phenotype, as some are better than others.
Indeed, Alrm-Gal4 comes on two chromosomes – we used BDSC #67032, which is on chromosome III and this is now clearly mentioned the Materials and Methods section.
- Figures 3 and 4: Labeling needs to remain consistent; Figure 3 "Glia-Gal4", Figure 4 "glia-gal4".
Thanks, done.
Reviewer #1 (Significance (Required)):
General Assessment: An interesting study on MMP function during an unusual type of neural development (axon pruning). Most of the MMP function appears to be in glia, although the MMP role in this context in unclear. The MMP function in the neurons being pruned is unexpected and even less clear. The study is somewhat poorly described in terse language lacking essential information, which gives the overall impression of a preliminary report.
Advance: Glial MMP function has been described for neuronal clearance mechanisms following injury. The main advance here is to describe a similar function during normal development. Audience: Developmental neuroscientists, MMP biologists, possibly schizophrenia clinician researchers
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
Neuropsychiatric conditions are often influenced by genetic factors. Schizophrenia is a complex mental disorder characterised by a mixture of hallucinations, delusions and disorganised thinking that causes lifelong problems in daily life. GWAS have identified a number of genes associated with the risk of developing schizophrenia, although genetic predisposition alone is not sufficient and additional environmental factors are required. In the current manuscript, the authors aim to exploit the strength of the Drosophila system to explore a link between schizophrenia-associated genes and neuronal remodelling during development. They focus on the mushroom body in the adult brain, where pronounced neuronal remodelling occurs during metamorphosis. To assess the potential role of the genes identified by the GWAS, they performed a targeted RNAi-based screen. They focus on the role of metalloproteases and find that they are required in neurons and in glia for the pruning of mushroom body axons. The study starts with a selection of 32 genes, 29 of which are listed (a bit hidden) in materials and methods and the identification of the Drosophila orthologs. The expression patterns of these genes in Kenyon cells are presented in Figure 1 - but unfortunately no information is given on who is expressed when
We apologize for the confusion. We attempted to keep Figure 1 simple but this resulted in the absence of critical information, as the reviewer suggests. We now include a Figure S1 that includes the entire heatmap of the dynamically expressed clusters I-IX with all the gene names. Additionally, we now augmented the information in Table 1 to include the screen phenotypes. Finally, Supplemental excel file 1, also included in our original submission, includes all the data, and is now better referred to throughout the text.
In a next step, Kenyon cell specific RNAi knockdown experiments are shown that identify a pruning phenotype for several genes. They demonstrate that Mmp2 (and similarly Mmp1) is also required in glia. Although Mmp2 was identified by neuronal RNAi-based knockdown, double knockdown experiments led the authors conclude that its primary function is in glia. The study emphasises the use of the advanced genetic model to understand complex human diseases. However, the paper does not go far enough in making use of the excellent genetics available. Basically, the report is about the identification of a few hits in a small RNAi screen, which is fine in itself, but leaves many questions unanswered. Do mmp1/2 mutants have a phenotype?
This is a very important question that cannot be answered, unfortunately. There is one published Mmp2 loss of function null allele which is lethal during pupal development (Page-MaCaw et al, 2003). Our previous data, using tissue specific (ts)CRISPR, suggested the involvement of Mmp2 in neurons for their remodeling (Meltzer et al, 2019). We therefore independently generated an Mmp2 germline mutant using CRISPR (harboring an indel resulting in a premature stop codon and predicted to encode a truncated, 77 amino-acid long protein), now described in Fig. S5A (and in the Materials and Methods). This allele is, as expected, unfortunately also lethal. We attempted to overcome lethality by generating MARCM (mosaic) clones in neurons, but as expected, because Mmp2 is largely secreted, there was no pruning defect phenotype (Fig. S5B-C). Unfortunately, it is not yet possible to generate glial clones. Additionally, available Mmp1 mutants are, sadly, also homozygous lethal. That said, in our revised manuscript we now include data demonstrating that expression of a dominant negative variant of Mmp1 inhibits pruning (Fig. 3J-K). We strengthened the evidence regarding the reliability of Mmp1 RNAi using an antibody mix (Fig. S4), and for Mmp2 – we refer to a manuscript that tested its efficiency (Harmansa et al., 2023). Lastly, we added new data using an additional RNAi line targeting Mmp2 from the VDRC collection (Fig. 3L).
Can the phenotype be rescued?
Unfortunately, without a viable mutant LOF phenotype, a rescue experiment is impossible. Regardless, in an attempt to rescue the RNAi phenotype, we designed and generated an RNAi-resistant Mmp2 overexpression transgene. Unfortunately, due to the destruction of our lab – several days after we received this transgenic line from Bestgene – this experiment is not included in the revision.
Does TIMP expression lead to similar phenotypes?
This is an interesting question which we addressed in our experiments but did not include in the text. Unfortunately, overexpression of TIMP did not have any effect on MB development. We are adding this figure here as Reviewer Figure 1, but we think that adding this information to the paper will not improve it for several reasons. The lack of phenotype by overexpression of Timp can result from a technical issue such as low expression or mislocalization of the protein, or a biological issue such as more complicated involvement of TIMP or other MMP inhibitors.
What is the temporal requirement for Mmp1/2?
This is an excellent suggestion, not an easy experiment, but one that we initiated, using a temperature sensitive Gal80 to control the expression of the RNAi only during metamorphosis. However, to the unfortunate destruction of our lab, this experiment was never completed.
What are the target proteins of Mmp2?
This is the million-dollar question – but unfortunately is beyond the scope of this short report.
Is Mmp2 still required when astrocyte motility is blocked? What is the morphology of glia after Mmp1/2 knockdown?
Thank you for this wonderful suggestion. We initiated two types of experiments using sparse labeling techniques (both MARCM and SPARC) to identify the morphology of single astrocytes in WT vs. MMP KD. However, these are complicated crosses that were not completed prior to the destruction of our lab.
Reviewer #2 (Significance (Required)):
The strength of the study is to identify a pruning phenotype after RNAi-based knockdown. The limitations is that this study is very superficial, it is the beginning of a paper. The initial claim to use Drosophila because to its advanced genetics is not met. The results section is shorter than the discussion.
While we agree with much of the reviewer’s statement this also relates to our general comment about “preliminary” type 1 and type 2 – True, this could be the beginning of a big paper and it would definitely be a more comprehensive and deep story. Most of the papers from my lab are indeed a 5 year endeavor. However, this short report (which is now longer, more detailed, and includes additional experiments) is a result of the work of an outstanding master’s student who came up with the idea for the project entirely by herself. Thus – given the data that she has acquired, and the fact that my lab will not continue to study MMPs or schizophrenia, the question needs to be whether the data supports the claims and whether this is an advance of science worthwhile of publication in a respectable journal. Our clear and decisive opinion is that the answer to that question is yes.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
In this work, Schuldiner and colleagues explore the role of Mmp1 and Mmp2 in neuronal remodeling in the mushroom body of Drosophila. Overall, this work is very interesting, but in its current form seems quite preliminary. The biggest limitation of the study is that single RNAi lines are used with no validation that the lines are working, despite the fact that Mmp antibodies are available as are endogenously tagged Mmp lines that could have been used to validate the genetic manipulations. Specific concerns are listed below.
We thank reviewer 3 for his generally positive assessment of our work and we now performed additional experiments to strengthen and validate the original RNAi findings – for specifics see our reply to the points below.
Major concerns 1) The scoring system for pruning of mushroom body neurons seems very variable, even in controls (where scoring can range from very mild to moderate), and it is very hard to assess from the images what one is looking at (rather than using our own judgment, we rely on the authors' words). It would be necessary to have better labeling and examples of what phenotypes are considered "mild", "severe", "wild type-like". It would also help to understand how phenotype assessment is guided by the overlap between the signals from TdTomato fluorescence and FasII stain.
We thank the reviewer for raising this point, that has also been highlighted by other reviewers in some form. First, we have generated Figure S3A-E to show examples of the ranking, which was now performed by two independent investigators. Second, ranking was performed by looking at TdTomato positive vertical axons that are outside of the αlobe (high FasII) – this is now better explained in the materials and methods. Additionally, while we would love to have a better scoring, and automatic, system – and even published a semi-automated scoring algorithm in Alyagor et al. 2018 (Figure 3O in the Alyagor paper), because the driver also labels vertical axons (ɑ/β) and because unpruned γ axons often express FasII, this quantification method does not always work. What we have done in previous cases, as we have also done here, is to provide independent ranking by two investigators and compare their ranking (Fig. S3F-G). Finally, we are working with our AI hub to develop automatic scoring systems that will not require human ranking – however this is beyond the scope for this manuscript.
2) The biggest limitations of the approach are that single RNAi lines are used to screen, with no accompanying validation of the tool (see above)
We agree. Unfortunately not all RNAis are “equal” and thus not all of them work. To support the RNAi data, we have better clarified previous experiments that demonstrate the importance of neuronal Mmp2 via tissue specific (ts) CRISPR (Meltzer, et al, 2019). Unfortunately, the Mmp2 null mutant that is available is lethal during pupal development (Page-MaCaw et al, 2003). We therefore independently generated an Mmp2 germline mutant using CRISPR (harboring an indel resulting in a premature stop codon and predicted to encode a truncated, 77 amino-acid long protein), now described in Fig. S5A (and in the Materials and Methods). This allele is, as expected, unfortunately also lethal. We attempted to overcome lethality by generating MARCM (mosaic) clones in neurons, but as expected, because Mmp2 is largely secreted, there was no pruning defect phenotype (Fig. S5B-C). Unfortunately, it is not yet possible to generate glial clones. Additionally, available Mmp1 mutants are, sadly, also homozygous lethal. That said, in our revised manuscript we now include data demonstrating that expression of a dominant negative variant of Mmp1 inhibits pruning (Fig. 3J-K). We strengthened the evidence regarding the reliability of Mmp1 RNAi using an antibody mix (Fig. S4), and for Mmp2 – we refer to a manuscript that tested its efficiency (Harmansa et al., 2023). Lastly, we added new data using an additional RNAi line targeting Mmp2 from the VDRC collection (Fig. 3L).
3) RNAi-based knockdown is used to infer epistatic information-this is not appropriate as epistasis experiments need to be done with null alleles to make firm conclusions. Additional concerns: ● Even with the same driver, knockdown efficiency for 2 different genes could be variable and dependent of the specific RNAi used. ● The comparison between drivers is even harder, as driver strength varies greatly. ● The knockdown efficiency drops with increasing numbers of RNAi used. ● The specific genotypes used for this experiment should be clarified, as it would be very important to ensure that the UAS dosage is equal across conditions.
We agree that RNAi is not optimal to assess epistasis. And indeed, we did not mean to claim epistasis relationship between Mmp1 and Mmp2, nor between neurons and glia. We now use better language to clarify this. To define epistatic relationships, the use of mutants would be required, unfortunately the use of nulls is not possible because they are lethal and secreted (thus not enabling mosaic analyses). We agree that increasing the number of RNAi lines is expected to reduce their efficiency – this is why it is even more significant when we see an increased defective phenotype in the double knockdown experiments. Finally, we totally agree about the genotype comment and apologize that it was erroneously omitted in the original submission– all of which have been now added (Table 2 in materials and methods).
4) To further deepen the rigor of this work, a few simple yet important things could have been done. First, it would be important to rule out that knocking down Mmps does not affect astrocyte numbers and health (could be assessed by counting numbers and observing their morphology). Also, the authors previously showed that astrocytes actively infiltrate the axon bundle prior to pruning to facilitate axon defasciculation and pruning (Marmor-Kollet et al., 2023). It would have provided an important insight to examine if astrocytes can infiltrate the axon bundle if Mmp2 and/or Mmp1 are knocked down.
Thank you for these wonderful suggestions. We embarked on a few experiments as detailed below, unfortunately these are complicated crosses that were not completed prior to the destruction of our lab. 1) We initiated two types of experiments using sparse labeling techniques (both MARCM and SPARC) to identify the morphology of single astrocytes in WT vs. MMP KD. 2) Testing astrocytic infiltrations requires three binary systems, we obtained and generated stocks required for these experiments, but these were prematurely terminated. 3) We initiated experiments to count the number of glial nuclei in the vicinity of the degenerating axonal lobe (at the onset of pruning). Preliminary experiments with a small n (3 controls, 4 Mmp1 RNAi, and 5 Mmp2 RNAi) suggest that the number of glial nuclei is not significantly different between these conditions.
Minor The introduction puts big emphasis on the role of glia, but then to narrows down candidate genes for the screen a γ-KCs transcriptional data set is used, and the initial screen is done via knockdown of those candidates in neurons (there is a disconnect between rationale and approach).
We totally agree with this reviewer which is why we now changed the paper to include both neuronal and glial loss-of-function screens. Figure 1 is now augmented with the glial data.
Rationale for looking into axon pruning and how that translates into insights about synaptic pruning defects in schizophrenia should be more clearly stated.
Indeed, our belief that synapse pruning and axon pruning share molecular mechanisms remains yet unproven. However, both are steps during neuronal remodeling, which has been previously implicated in schizophrenia. That said, we now added an additional disclaimer to acknowledge the limitation of our findings in the context of human disease and synapse elimination (lines 275-279).
Figure 1C: data visualization for this heat map should be improved. Parts of the data are faded, and the differences between gene clusters are unclear.
We apologize for the incomplete figure. We did not want to overload the figure with data, which is why we are showing only the important clusters and did not include gene names. To keep the figure simple, but at the same time provide the complete information, we now include the full data in Fig. S1 (that includes the original heatmap with all the dynamic clusters I-IX, and including all the gene names). For the full raw data, including non-dynamic clusters, the reader is referred to look in Supplemental excel file 1. We hope this provides the clarity that this reviewer rightfully asks for.
Reviewer #3 (Significance (Required)):
In this work, Schuldiner and colleagues explore the role of Mmp1 and Mmp2 in neuronal remodeling in the mushroom body of Drosophila. Overall, this work is very interesting, but in its current form seems quite preliminary. The biggest limitation of the study is that single RNAi lines are used with no validation that the lines are working, despite the fact that Mmp antibodies are available as are endogenously tagged Mmp lines that could have been used to validate the genetic manipulations.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this work, Schuldiner and colleagues explore the role of Mmp1 and Mmp2 in neuronal remodeling in the mushroom body of Drosophila. Overall, this work is very interesting, but in its current form seems quite preliminary. The biggest limitation of the study is that single RNAi lines are used with no validation that the lines are working, despite the fact that Mmp antibodies are available as are endogenously tagged Mmp lines that could have been used to validate the genetic manipulations. Specific concerns are listed below.
Major concerns
Minor
The introduction puts big emphasis on the role of glia, but then to narrows down candidate genes for the screen a γ-KCs transcriptional data set is used, and the initial screen is done via knockdown of those candidates in neurons (there is a disconnect between rationale and approach).
Rationale for looking into axon pruning and how that translates into insights about synaptic pruning defects in schizophrenia should be more clearly stated.
Figure 1C: data visualization for this heat map should be improved. Parts of the data are faded, and the differences between gene clusters are unclear.
In this work, Schuldiner and colleagues explore the role of Mmp1 and Mmp2 in neuronal remodeling in the mushroom body of Drosophila. Overall, this work is very interesting, but in its current form seems quite preliminary. The biggest limitation of the study is that single RNAi lines are used with no validation that the lines are working, despite the fact that Mmp antibodies are available as are endogenously tagged Mmp lines that could have been used to validate the genetic manipulations.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Neuropsychiatric conditions are often influenced by genetic factors. Schizophrenia is a complex mental disorder characterised by a mixture of hallucinations, delusions and disorganised thinking that causes lifelong problems in daily life. GWAS have identified a number of genes associated with the risk of developing schizophrenia, although genetic predisposition alone is not sufficient and additional environmental factors are required.
In the current manuscript, the authors aim to exploit the strength of the Drosophila system to explore a link between schizophrenia-associated genes and neuronal remodelling during development. They focus on the mushroom body in the adult brain, where pronounced neuronal remodelling occurs during metamorphosis. To assess the potential role of the genes identified by the GWAS, they performed a targeted RNAi-based screen. They focus on the role of metalloproteases and find that they are required in neurons and in glia for the pruning of mushroom body axons.
The study starts with a selection of 32 genes, 29 of which are listed (a bit hidden) in materials and methods and the identification of the Drosophila orthologs. The expression patterns of these genes in Kenyon cells are presented in Figure 1 - but unfortunately no information is given on who is expressed when. In a next step, Kenyon cell specific RNAi knockdown experiments are shown that identify a pruning phenotype for several genes. They demonstrate that Mmp2 (and similarly Mmp1) is also required in glia. Although Mmp2 was identified by neuronal RNAi-based knockdown, double knockdown experiments led the authors conclude that its primary function is in glia.
The study emphasises the use of the advanced genetic model to understand complex human diseases. However, the paper does not go far enough in making use of the excellent genetics available. Basically, the report is about the identification of a few hits in a small RNAi screen, which is fine in itself, but leaves many questions unanswered. Do mmp1/2 mutants have a phenotype? Can the phenotype be rescued? Does TIMP expression lead to similar phenotypes? What is the temporal requirement for Mmp1/2? What are the target proteins of Mmp2? Is Mmp2 still required when astrocyte motility is blocked? What is the morphology of glia after Mmp1/2 knockdown?
The strength of the study is to identify a pruning phenotype after RNAi-based knockdown. The limitations is that this study is very superficial, it is the beginning of a paper. The initial claim to use Drosophila because to its advanced genetics is not met. The results section is shorter than the discussion.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
This study reports a role for matrix metalloproteinases (MMPs) in the developmental pruning of gamma Kenyon cells (KCs) in the fruit fly Mushroom Body during larval-pupal metamorphosis. The authors show through gene expression studies that MMP genes are upregulated in late larval stages as part of the early program for this type of neuronal pruning. They show through cell-targeted RNAi studies of both secreted MMP-1 and membrane-anchored MMP-2, that both genes are required in glial cells and to a lesser extent within KCs. The authors show that MMP secreted from glial is required for normal levels of Mushroom Body developmental neuronal pruning. They mention that MMP genes have been identified in schizophrenic patient screens in patients, and that perhaps a comparable pruning mechanism could be involved in the loss of grey matter (loss of synapses) in patients. The authors propose that MMP levels may be a potential therapeutic marker in the future.
Major Comments:
Overall, the work is of a reasonable standard, but very preliminary. The study lacks the substance to completely convince me of any of the results. There is SUBSTANTIAL work that needs to be done to make this publishable. There are a lot of writing mistakes; so many that I do not list them in detail here. The references citations are fairly old, but I do not list update replacements here. The text is very brief, and the overall writing needs to include significantly more description and detail. This is evident in all aspects of the manuscript, but especially notable in the Methods and Figure Legends. None of the Figure Legends include full genotypes of any of the fly lines, and these full fly lines are also not included in the Methods. This is vital to compare the experimental lines to the controls. Major points are listed below:
Minor Comments:
General Assessment: An interesting study on MMP function during an unusual type of neural development (axon pruning). Most of the MMP function appears to be in glia, although the MMP role in this context in unclear. The MMP function in the neurons being pruned is unexpected and even less clear. The study is somewhat poorly described in terse language lacking essential information, which gives the overall impression of a preliminary report.
Advance: Glial MMP function has been described for neuronal clearance mechanisms following injury. The main advance here is to describe a similar function during normal development.
Audience: Developmental neuroscientists, MMP biologists, possibly schizophrenia clinician researchers
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
We thank the reviewers for providing us the opportunity to revise our manuscript titled “Identifying regulators of associative learning using a protein-labelling approach in C. elegans.” We appreciate the insightful feedback that we received to improve this work. In response, we have extensively revised the manuscript with the following changes: we have (1) clarified the criteria used for selecting candidate genes for behavioural testing, presenting additional data from ‘strong’ hits identified in multiple biological replicates (now testing 26 candidates, previously 17), (2) expanded our discussion of the functional relevance of validated hits, including providing new tissue-specific and neuron class-specific analyses, and (3) improved the presentation of our data, including visualising networks identified in the ‘learning proteome’, to better highlight the significance of our findings. We also substantially revised the text to indicate our attempts to address limitations related to background noise in the proteomic data and outlined potential refinements for future studies. All revisions are clearly marked in the manuscript in red font. A detailed, point-by-point response to each comment is provided below.
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
Summary:
Rahmani et al., utilize the TurboID method to characterize the global proteome changes in the worm's nervous system induced by a salt-based associative learning paradigm. Altogether, Rahmani et al., uncover 706 proteins that are tagged by the TurboID method specifically in samples extracted from worms that underwent the memory inducing protocol. Next, the authors conduct a gene enrichment analysis that implicates specific molecular pathways in salt-associative learning, such as MAP-kinase and cAMP-mediated pathways. The authors then screen a representative group of the hits from the proteome analysis. The authors find that mutants of candidate genes from the MAP-kinase pathway, namely dlk-1 and uev-3, do not affect the performance in the learning paradigm. Instead multiple acetylcholine signaling mutants significantly affected the performance in the associative memory assay, e.g., acc-1, acc-3, gar-1, and lgc-46. Finally, the authors demonstrate that the acetylcholine signaling mutants did not exhibit a phenotype in similar but different conditioning paradigms, such as aversive salt-conditioning or appetitive odor conditioning, suggesting their effect is specific to appetitive salt conditioning.
Major comments:
Since most of the control data in this assay in this study is very close to 1, it strongly suggests that the CI data is not normally distributed and therefore 2-way ANOVA is expected to give skewed results.
I am aware this is a common mistake and I also anticipate that most conclusions will still hold also under a more fitting statistical test.
We appreciate the point raised by Reviewer 1 and understand the importance of performing the correct statistical tests.
The statistical tests used in this study were chosen since parametric tests, particularly ANOVA tests to assess differences between multiple groups, are commonly used to assess behaviour in the C. elegans learning and memory field. Below is a summary of the tests used by studies that perform similar behavioural tests cited in this work, as examples:
Table 1 | A summary for the statistical tests performed by similar studies for chemotaxis assay data. References (listed in the leftmost column) were observed to (A) use parametric tests only or (B) performed either a parametric or non-parametric test on each chemotaxis assay dataset depending on whether the data passed a normality test. Listings for ANOVA tests are in bold to demonstrate their common use in the C. elegans learning and memory field.
Reference
Parametric test/s used in the reference
Non-parametric test/s used in the reference
Beets et al., 2020
Two-way ANOVA
None
Hiroki & Iino 2022
One-way ANOVA
None
Hiroki et al., 2022
One-way ANOVA
None
Hukema et al., 2006
T-tests
None
Hukema et al., Learn. Mem. 2008
T-tests
None
Jang et al., 2019
ANOVA
None
Kitazono et al., 2017
Two-way ANOVA and t-tests
None
Lans et al., 2004
One-way ANOVA
None
Lim et al., 2018
Two-way ANOVA
Wilcoxon rank sum test adjusted with the Benjamini–Hochberg method
Lin et al., 2010
Two-way or three-way ANOVA
None
Nagashima et al., 2019
One-way ANOVA
None
Ohno et al., 2014
None
Sakai et al., 2017
One-way ANOVA or t-tests
None
Stein & Murphy 2014
Two-way ANOVA and t-tests
None
Tang et al., 2023
One-way ANOVA or t-tests
None
Tomioka et al., 2006
T tests
None
Watteyne et al., 2020
One-way ANOVA
Two-sided Kruskal–Wallis
We note Reviewer 1's concern that this may stem from a common mistake. As stated, Two-way ANOVA generally relies on normally distributed data. We used GraphPad Prism to perform the Shapiro-Wilk normality test on our chemotaxis assay data as it is generally appropriate for sample sizes Table 2 | Shapiro-Wilk normality test results for chemotaxis assay data in Figure S8C. Chemotaxis assay data was generated to assess salt associative learning capacity for wild-type (WT) versus lgc-46(-) mutant C. elegans. Three experimental groups were prepared for each C. elegans strain (naïve, high-salt control, and trained). From top-to-bottom, the data below displays the ‘W’ value, ‘P value’, a binary yes/no for whether the data passes the Shapiro-Wilk normality test, and a ‘P value summary’ (ns = non-significant). W values measure the similarity between a normal distribution and the chemotaxis assay data. Data is considered normal in the Shapiro-Wilk normality test when a W value is near 1.0 and the null hypothesis is not rejected (i.e., P value > 0.05).*
WT naïve
WT high-salt control
WT trained
lgc-46 naïve
lgc-46 high-salt control
lgc-46 trained
W
0.9196
0.9114
0.8926
0.8334
0.8151
0.8769
P value
0.5272
0.4758
0.3705
0.1475
0.1070
0.2954
Passed normality test (alpha=0.05)?
Yes
Yes
Yes
Yes
Yes
Yes
P value summary
ns
ns
ns
ns
ns
ns
The manuscript now includes the use of the Shapiro-Wilk normality test to assess chemotaxis assay data before using two-way ANOVA on page 51.
Nevertheless an appropriate statistical analysis should be performed. Since I assume the authors would wish to take into consideration both the different conditions and biological repeats, I can suggest two options:
Using a custom bootstrapping approach. We thank Reviewer 1 for suggesting these two options. We carefully considered both approaches and consulted with the in-house statistician at our institution (Dr Pawel Skuza, Flinders University) for expert advice to guide our decision. In summary:
Generalised linear mixed models: Generalised linear mixed models (GLMMs) are generally most appropriate for nested/hierarchal data. However, our chemotaxis assay data does not exhibit such nesting. Each biological replicate (N) consists of three technical replicates, which are averaged to yield a single chemotaxis index per N. Our statistical comparisons are based solely on these averaged values across experimental groups, making GLMMs less applicable in this context.
We thoroughly evaluated performing the power analysis: however, this is typically performed with the assumption that an N = 1 represents a singular individual/person. An N =1 in this study is one biological replicate that includes hundreds of worms, which is why it is not typically employed in our field for this type of behavioural test.
Considering these factors, we have opted to continue using a two-way ANOVA for our statistical analysis. This choice aligns with recent publications that employ similar experimental designs and data structures. Crucially, we have verified that our data meet the assumptions of normality, addressing key concerns regarding the suitability of parametric testing. We believe this approach is sufficiently rigorous to support our main conclusions. This rationale is now outlined on page 51.
To be fully transparent, our aim is to present differences between wild-type and mutant strains that are clearly visible in the graphical data, such that the choice of statistical test does not become a limiting factor in interpreting biological relevance. We hope this rationale is understandable, and we sincerely appreciate the reviewer’s comment and the opportunity to clarify our analytical approach.
We hope that Reviewer 1 will appreciate these considerations as sufficient justification to retain the statistical tests used in the original manuscript. Nevertheless, to constructively address this comment, we have performed the following revisions:
Reference
N used in the study for chemotaxis assay data
Beets et al., 2020
8
Hiroki & Iino 2022
5-8
Hiroki et al., 2022
6-7
Hukema et al., 2006
≥ 4
Hukema et al., Learn. Mem. 2008
≥ 4
Jang et al., 2019
≥ 4
Kitazono et al., 2017
≥ 4
Kauffman et al., 2010
≥ 3
Kauffman et al., J. Vis. Exp. 2011
≥ 3
Lans et al., 2004
2
Lim et al., 2018
2-4
Lin et al., 2010
≥ 4
Nagashima et al., 2019
≥ 7
Ohno et al., 2014
≥ 11
Sakai et al., 2017
≥ 4
Stein & Murphy 2014
3-5
Tang et al., 2023
≥ 9
Watteyne et al., 2020
≥ 10
__Grouped presentation of behavioural data: __We now present all behavioural data by grouping genotypes tested within the same biological replicate, including wild-type controls, rather than combining genotypes tested separately. This ensures that each graph displays data from genotypes sharing the same N, also an important consideration for performing parametric tests. Accordingly, we re-performed statistical analyses using this reduced Nfor relevant graphs. As anticipated, this rendered some comparisons non-significant. All statistical comparisons are clearly indicated on each graph. Improved clarity of figure legends: __We revised figure legends for __Figures 5, 6, S7, S8, & S9 to make clear how many biological replicates have been performed for each genotype by adding N numbers for each genotype in all figures.
The authors use the phrasing "a non-significant trend", I find such claims uninterpretable and should be avoided. Examples: Page 16. Line 7 and Page 18, line 16.
This is an important point. While we were not able to find the specific phrasing "a non-significant trend" from this comment in the original manuscript, we acknowledge that referring to a phenotype as both a trend and non-significant may confuse readers, which was originally stated in the manuscript in two locations.
The main text has been revised on pages 27 & 28 when describing comparisons between trained groups between two C. elegans lines, by removing mentions of trends and retaining descriptions of non-significance.
Neuron-specific analysis and rescue of mutants:
Throughout the study the authors avoid focusing on specific neurons. This is understandable as the authors aim at a systems biology approach, however, in my view this limits the impact of the study. I am aware that the proteome changes analyzed in this study were extracted from a pan neuronally expressed TurboID. Yet, neuron-specific changes may nevertheless be found. For example, running the protein lists from Table S2, in the Gene enrichment tool of wormbase, I found, across several biological replicates, enrichment for the NSM, CAN and RIG neurons. A more careful analysis may uncover specific neurons that take part in this associative memory paradigm. In addition, analysis of the overlap in expression of the final gene list in different neurons, comparing them, looking for overlap and connectivity, would also help to direct towards specific circuits.
This is an important and useful suggestion. We appreciate the benefit in exploring the data from this study from a neuron class-specific lens, in addition to the systems-level analyses already presented.
The WormBase gene enrichment tool is indeed valuable for broad transcriptomic analyses (the findings from utilising this tool are now on page 16); however, its use of Anatomy Ontology (AO) terms also contains annotations from more abundant non-neuronal tissues in the worm. To strengthen our analysis and complement the Wormbase tool, we also used the CeNGEN database as suggested by Reviewer 3 Major Comment 1 (Taylor et al., 2021), which uses single cell RNA-Seq data to profile gene expression across the C. elegans nervous system. We input our learning proteome data into CeNGEN as a systemic analysis, identifying neurons highly represented by the learning proteome (on pages 16-20). To do this, we specifically compared genes/proteins from high-salt control worms and trained worms to identify potential neurons that may be involved in this learning paradigm. Briefly, we found:
To further address the reviewer’s suggestion, we examined the overlap in expression patterns of the validated learning-associated genes acc-1, acc-3, lgc-46, kin-2, and F46H5.3 across the neuron classes above, using the CeNGEN database. This was done to explore potential neuron classes in which these regulators may act in to regulate learning. This analysis revealed both shared and distinct expression profiles, suggesting potential functional connectivity or co-regulation among subsets of neurons. To summarise, we found:
OPTIONAL: A rescue of the phenotype of the mutants by re-expression of the gene is missing, this makes sure to avoid false-positive results coming from background mutations. For example, a pan neuronal or endogenous promoter rescue would help the authors to substantiate their claims, this can be done for the most promising genes. The ideal experiment would be a neuron-specific rescue but this can be saved for future works.
We appreciate this suggestion and recognise its potential to strengthen our manuscript. In response, we made many attempts to generate pan-neuronal and endogenous promoter re-expression lines. However, we faced several technical issues in transgenic line generation, including poor survival following microinjection likely due to protein overexpression toxicity (e.g., C30G12.6, F46H5.3), and reduced animal viability for chemotaxis assays, potentially linked to transgene-related reproductive defects (e.g., ACC-1). As we have previously successfully generated dozens of transgenic lines in past work (e.g. Chew et al., Neuron 2018; Chew et al., Phil Trans B 2018; Gadenne/Chew et al., Life Science Alliance 2022), we believe the failure to produce most of these lines is not likely due to technical limitations. For transparency, these observations have been included in the discussion section of the manuscript on pages 39 & 40 as considerations for future troubleshooting.
Fortunately, we were able to generate a pan-neuronal promoter line for KIN-2 that has been tested and included in the revised manuscript. This new data is shown in Figure 5B __and described on __pages 23 & 24. Briefly, this shows that pan-neuronal expression of KIN-2 from the ce179 mutant allele is sufficient to reproduce the enhanced learning phenotype observed in kin-2(ce179) animals, confirming the role of KIN-2 in gustatory learning.
To address the potential involvement of background mutations (also indicated by Reviewer 4 under ‘cross-commenting’), we have also performed experiments with backcrossed versions of several mutants. These experiments aimed to confirm that salt associative learning phenotypes are due to the expected mutation. Namely, we assessed kin-2(ce179) mutants that had been backcrossed previously by another laboratory, as well as C30G12.6(-) and F46H5.3(-) animals backcrossed in this study. Although not all backcrossed mutants retained their original phenotype (i.e., C30G12.6) (Figure 6D, a newly added figure), we found that backcrossed versions of KIN-2 and F46H5.3 both robustly showed enhanced learning (Figures 5A & 6B). This is described in the text on pages 23-26.
__Minor comments: __
This is an important point: We validated our biotin tagging method prior to mass spectrometry by comparing ‘no biotin’ and ‘biotin’ groups. This is shown in Figure S1 in the revised manuscript, which includes a western blot comparing untreated and biotin treated animals that are non-transgenic or expressing TurboID. As expected, by comparing biotinylated protein signal for untreated and treated lanes within each line, biotin treatment increased the signal 1.30-fold for non-transgenic and 1.70-fold for TurboID C. elegans. This is described on __page 8 __of the revised manuscript.
To clarify, for mass spectrometry experiments, we tested a no-TurboID (non-transgenic) control, but did not perform a no-biotin control. We included the following four groups: (1) No-TurboID ‘control’ (2) No-TurboID ‘trained’, (3) pan-neuronal TurboID ‘control’ and (4) pan-neuronal TurboID ‘trained’, where trained versus control refers to whether ‘no salt’ was used as the conditioned stimulus or not, respectively (illustrated in Figure 1A). Due to the complexity of the learning assay (which involves multiple washes and handling steps, including a critical step where biotin is added during the conditioning period), and the need to collect sufficient numbers of worms for protein extraction (>3,000 worms per experimental group), adding ‘no-biotin’ controls would have doubled the number of experimental groups, which we considered unfeasible for practical reasons. This is explained on __pages 8 & 9 __of the revised manuscript.
Also, it was unclear which exact samples were tested per replicate. In Page 9, Lines 17-18: "For all replicates, we determined that biotinylated proteins could be observed ...", But in Page 8, Line 24 : "We then isolated proteins from ... worms per group for both 'control' and 'trained' groups,... some of which were probed via western blotting to confirm the presence of biotinylated proteins".
Thank you for pointing out these unclear statements: We have clarified the experimental groups used for mass spectrometry experiments as detailed in the response above on pages 8 &____ 9. In addition, western blots corresponding to each biological replicate of mass spectrometry data described in the main text on page 10 and have been added to the revised manuscript (as Figure S3). These western blots compare biotinylation signal for proteins extracted from (1) No-TurboID ‘control’ (2) No-TurboID ‘trained’, (3) pan-neuronal TurboID ‘control’ and (4) pan-neuronal TurboID ‘trained’. These blots function to confirm that there were biotinylated proteins in TurboID samples, before enrichment by streptavidin-mediated pull-down for mass spectrometry.
OPTIONAL: include the fold changes of biotinylated proteins of all the ones that were tested. Similar to Figure 1.C.
This is an excellent suggestion. As recommended by the reviewer, we have included fold-changes for biotinylated protein levels between high-salt control and trained groups (on pages 9 & 10 for replicate #1 and in __Table S2 __for replicates #2-5). This was done by measuring protein levels in whole lanes for each experimental group per biological replicate within western blots (__Figure 1C __for replicate #1 and __Figure S3 __for replicates #2-5) of protein samples generated for mass spectrometry (N = 5).
Figure 2 does not add much to the reader, it can be summarized in the text, as the fraction of proteins enriched for specific cellular compartments.
As noted in cross-comment response to Reviewer 4, there were typos in the original figure references, we have corrected them above. Essentially, this comment is referring to Figure 2.
We appreciate this feedback from Reviewer 1. We agree that the original __Figure 2 __functions as a visual summary from analysis of the learning proteome at the subcellular compartment level. However, it also serves to highlight the following:
Many of these candidates could not be assessed by learning assay using single mutants since related mutations are lethal or substantially affect locomotion. These networks therefore highlight the benefit in using strategies like TurboID to study learning. We have chosen to retain this figure, moving it to the supplementary material as Figure S4 in the revised manuscript, as suggested.
OPTIONAL- I would suggest the authors to mark in a pathway summary figure similar to Figure 3 (originally written as Figure 4) the results from the behavior assay of the genetic screen. This would allow the reader to better get the bigger picture and to connect to the systemic approach taken in Figures 2 and 3.
We think this is a fantastic suggestion and thank Reviewer 1 for this input. In the revised manuscript, we have added Figure 7, which summarises the tested candidates that displayed an effect on learning, mapped onto potential molecular pathways derived from networks in the learning proteome. This figure provides a visual framework linking the behavioural outcomes to the network context. This is described in the main text on pages 32-33.
Typo in Figure 3: the circle of PPM1: The blue right circle half is bigger than the left one.
We thank the Reviewer for noticing this, the node size for PPM-1.A has been corrected in what is now Figure 2 in the revised work.
Unclarity in the discussions. In the discussion Page 24, Line 14, the authors raise this question: "why are the proteins we identified not general learning regulators?. The phrasing and logic of the argumentation of the possible answers was hard to follow. - Can you clarify?
We appreciate this feedback in terms of unclarity, as we strive to explain the data as clearly and transparently as possible. Our goal in this paragraph was to discuss why some candidates were seen to only affect salt associative learning, as opposed to showing effects in multiple learning paradigms (i.e., which we were defining as a ‘general learning regulator’). We have adjusted the wording in several places in this paragraph now on pages 36 & 37 to address this comment. We hope the rephrased paragraph provides sufficient rationalisation for the discussion regarding our selection strategy used to isolate our protein list of potential learning regulators, and its potential limitations.
***Cross-Commenting** *
Firstly, we would like to express our appreciation for the opportunity for reviewers to cross-comment on feedback from other reviewers. We believe this is an excellent feature of the peer review process, and we are grateful to the reviewers for their thoughtful engagement and collaborative input.
I would like to thank Reviewer #4 for the great cross comment summary, I find it accurate and helpful.
I also would like to thank Reviewer #4 for spotting the typos in my minor comments, their page and figure numbers are the correct ones.
We have corrected these typos in the relevant comments, and have responded to them accordingly.
Small comment on common point 1 - My feeling is that it is challanging to do quantitative mass spectrometry, especially with TurboID. In general, the nature of MS data is that it hints towards a direction but a followup validation work is required in order to assess it. For example, I am not surprised that the fraction of repeats a hit appeared in does not predict well whether this hit would be validated behavioraly. Given these limitations, I find the authors' approach reasonable.
We thank Reviewer 1 for this positive and thoughtful feedback. We also appreciate Reviewer 4’s comment regarding quantitative mass spectrometry and have addressed this in detail below (see response to Reviewer 4). However, we agree with Reviewer 1 that there are practical challenges to performing quantitative mass spectrometry with TurboID, primarily due to the enrichment for biotinylated proteins that is a key feature of the sample preparation process.
Importantly, we whole-heartedly agree with Reviewer 1’s statement that “In general, the nature of MS data is that it hints towards a direction but a follow-up validation work is required in order to assess it”. This is the core of our approach: however, we appreciate that there are limitations to a qualitative ‘absent/present’ approach. We have addressed some of these limitations by clarifying the criteria used for selecting candidate genes, based additionally on the presence of the candidate in multiple biological replicates (categorised as ‘strong’ hits). Based on this method, we were able to validate the role of several novel learning regulators (Figures 5, 6, & S7). We sincerely hope that this manuscript can function as a direction for future research, as suggested by this Reviewer.
I also would like to highlight this major comment from reviewer 4:
"In Experimental Procedures, authors state that they excluded data in which naive or control groups showed average CI 0.5499 for N2 (page 36, lines 5-7). "
This threshold seems arbitrary to me too, and it requires the clarifications requested by reviewer 4.
As detailed in our response to Reviewer 4, Major Comment 2, data were excluded only in rare cases, specifically when N2 worms failed to show strong salt attraction prior to training, or when trained N2 worms did not exhibit the expected behavioural difference compared to untrained controls – this can largely be attributed to clear contamination or over-population issues, which are visible prior to assessing CTX plates and counting chemotaxis indices.
These criteria were initially established to provide an objective threshold for excluding biological replicates, particularly when planning to assay a large number of genetic mutants. However, after extensive testing across many replicates, we found that N2 worms (that were not starved, or not contaminated) consistently displayed the expected phenotype, rendering these thresholds unnecessary. We acknowledge that emphasizing these criteria may have been misleading, and have therefore removed them from page 50 in the revised manuscript to avoid confusion and ensure clarity.
Reviewer #1 (Significance (Required)):
This study does a great job to effectively utilize the TurboID technique to identify new pathways implicated in salt-associative learning in C. elegans. This technique was used in C. elegans before, but not in this context. The salt-associative memory induced proteome list is a valuable resource that will help future studies on associative memory in worms. Some of the implicated molecular pathways were found before to be involved in memory in worms like cAMP, as correctly referenced in the manuscript. The implication of the acetylcholine pathway is novel for C. elgeans, to the best of my knowledge. The finding that the uncovered genes are specifically required for salt associative memory and not for other memory assays is also interesting.
However overall I find the impact of this study limited. The premise of this work is to use the Turbo-ID method to conduct a systems analysis of the proteomic changes. The work starts by conducting network analysis and gene enrichment which fit a systemic approach. However, since the authors find that ~30% of the tested hits affect the phenotype, and since only 17/706 proteins were assessed, it is challenging to draw conclusive broad systemic claims. Alternatively, the authors could have focused on the positive hits, and understand them better, find the specific circuits where these genes act. This could have increased the impact of the work. Since neither of these two options are satisfied, I view this work as solid, but not wide in its impact and therefore estimate the audience of this study would be more specialized.
My expertise is in C. elegans behavior, genetics, and neuronal activity, programming and machine learning.
We thank the Reviewer for these comments and appreciate the recognition of the value of the proteomic dataset and the identification of novel molecular pathways, including the acetylcholine pathway, as well as the specificity of the uncovered genes to salt-associative memory.
Regarding the reviewer’s concern about the overall impact and scope of the study, we respectfully offer the following clarification. Our aim was to establish a systems-level approach for investigating learning-related proteomic changes using TurboID, and we acknowledge that only a subset of the identified proteins was experimentally tested (now 26/706 proteins in the revised manuscript). Although only five of the tested single gene mutants showed a robust learning phenotype in the revised work (after backcrossing, more stringent candidate selection, improved statistical analysis in addressing reviewer comments), our proteomic data provides us a unique opportunity to define these candidates within protein-protein networks (as illustrated in Figure 7). Importantly, our functional testing focused on single-gene mutants, which may not reveal phenotypes for genes that act redundantly (now mentioned on pages 28-30). This limitation is inherent to many genetic screens and highlights the value of our proteomic dataset, which enables the identification of broader protein-protein interaction networks and molecular pathways potentially involved in learning.
To support this systems-level perspective, we have added Figure 7, which visually integrates the tested candidates into molecular pathways derived from the learning proteome for learning regulators KIN-2 and F46H5.3. We also emphasise more explicitly in the text (on pages 32-33) the value of our approach by highlighting the functional protein networks that can be derived from our proteomics dataset.
We fully acknowledge that the use of TurboID across all neurons limits the resolution needed to pinpoint individual neuron contributions, and understand the benefit in further experiments to explore specific circuits. Many circuits required for salt sensing and salt-based learning are highly explored in the literature and defined explicitly (see Rahmani & Chew, 2021), so our intention was to complement the existing literature by exploring the protein-protein networks involved in learning, rather than on neuron-neuron connectivity. However, we recognise the benefit in integrating circuit-level analyses, given that our proteomic data suggests hundreds of candidates potentially involved in learning. While validating each of these candidates is beyond the scope of the current study, we have taken steps to suggest candidate neurons/circuits by incorporating tissue enrichment analyses and single-cell transcriptomic data (Table S7 & Figure 4). These additions highlight neuron classes of interest and suggest possible circuits relevant to learning.
We hope this clarification helps convey the intended scope and contribution of our study. We also believe that the revisions made in response to Reviewer 1’s feedback have strengthened the manuscript and enhanced its significance within the field.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
__Summary: __
In this study by Rahmani in colleagues, the authors sought to define the "learning proteome" for a gustatory associative learning paradigm in C. elegans. Using a cytoplasmic TurboID expressed under the control of a pan-neuronal promoter, the authors labeled proteins during the training portion of the paradigm, followed by proteomics analysis. This approach revealed hundreds of proteins potentially involved in learning, which the authors describe using gene ontology and pathways analysis. The authors performed functional characterization of some of these genes for their requirement in learning using the same paradigm. They also compared the requirement for these genes across various learning paradigms, and found that most hits they characterized appear to be specifically required for the training paradigm used for generating the "learning proteome".
Major Comments:
We also agree with Reviewer 2 that the overlap between individual candidate hits is low between biological replicates; the inclusion of Figure S2 __in the original manuscript serves to highlight this limitation. However, it is also important to consider that there is notable overlap for whole molecular pathways between biological replicates of mass spectrometry data as shown in __Figure 2 __in the revised manuscript (this consideration is now mentioned on __pages 13-14). We have included Figure 3 to illustrate representation for two metabolic processes across several biological replicates normally indispensable to animal health, as an example to provide additional visual aid for the overlap between replicates of mass spectrometry. We provide this figure (described on pages 13 & 15) to demonstrate the strength of our approach in that it can detect candidates not easily assessable by conventional forward or reverse genetic screens.
We also appreciate the opportunity to explain our approach. The criteria of “at least one unique peptide” was chosen based on a previous work for which we adapted for this manuscript (Prikas et al., 2020). It was not intended to inflate the number of hits but rather to ensure sensitivity in detecting low-abundance neuronal proteins. We have clarified this in our Methods (page 46).
The "hits" that the authors chose to functionally characterize do not seem like strong candidate hits based on the proteomics data that they generated. Indeed, most of the hits are present in a single, or at most 2, biological replicate. It is unclear as to why the strongest hits were not characterized, which if mutant strains are publicly available, would not be a difficult experiment to perform.
We thank the reviewer for this important suggestion. To address this, we have described two molecular pathways with multiple components that appear in more than one biological replicate of mass spectrometry data in Figure 3 (main text on page 13). In addition, we have included __Figures 6 & S7 __where 9 additional single mutants corresponding to candidates in three or more biological replicates of mass spectrometry were tested for salt associative learning. Briefly, we found the following (number of replicates that a protein was unique to TurboID trained animals is in brackets):
We thank Reviewer 2 for this suggestion – we agree that it would have been ideal to have additional evidence suggesting that changes in candidate protein levels are associated directly with learning. Ideally, we would have explored this aspect further; however, as outlined in response to Reviewer 1 Major Comment 2 (OPTIONAL), this was not feasible within the scope of the current study due to several practical challenges. Specifically, we attempted to generate pan-neuronal and endogenous promoter rescue lines for several candidates, but encountered significant challenges, including poor survival post-microinjection (likely due to protein overexpression toxicity) and reduced viability for behavioural assays, potentially linked to transgene-related reproductive defects. This information is now described on pages 39 & 40 of the revised work.
To address these limitations, we performed additional behavioural experiments where possible. We successfully generated a pan-neuronal promoter line for kin-2, which was tested and included in the revised manuscript (Figure 5B, pages 30 & 31). In addition, to confirm that observed learning phenotypes were due to the expected mutations and not background effects, we conducted experiments using backcrossed versions of several mutant lines as suggested by Reviewer 4 Cross Comment 3 (Figure 6, pages 23-24 & 24-26). Briefly, this shows that pan-neuronal expression of KIN-2 from the ce179 mutant allele is sufficient to repeat the enhanced learning phenotype observed in backcrossed kin-2(ce179) animals, providing additional evidence that the identified hits are required for learning. We also confirmed that F46H5.3 modulates salt associative learning, given both non-backcrossed and backcrossed F46H5.3(-) mutants display a learning enhancement phenotype. The revised text now describes this data on the page numbers mentioned above.
Minor Comments:
The authors highlight that the proteins they discover seem to function uniquely in their gustatory associative paradigm, but this is not completely accurate. kin-2, which they characterize in figure 4, is required for positive butanone association (the authors even say as much in the manuscript) in Stein and Murphy, 2014. We appreciate this correction and thank the Reviewer for pointing this out. We have amended the wording appropriately on page 31 to clarify our meaning.
“Although kin-2(ce179) mutants were not shown to impact salt aversive learning, they have been reported previously to display impaired intermediate-term memory (but intact learning and short-term memory) for butanone appetitive learning (Stein and Murphy, 2014).”*
Reviewer #2 (Significance (Required)):
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
__Summary: __
In the manuscript titled "Identifying regulators of associative learning using a protein-labelling approach in C. elegans" the authors attempted to generate a snapshot of the proteomic changes that happen in the C. elegans nervous system during learning and memory formation. They employed the TurboID-based protein labeling method to identify the proteins that are uniquely found in samples that underwent training to associate no-salt with food, and consequently exhibited lower attraction to high salt in a chemotaxis assay. Using this system they obtained a list of target proteins that included proteins represented in molecular pathways previously implicated in associative learning. The authors then further validated some of the hits from the assay by testing single gene mutants for effects on learning and memory formation.
Major Comments:
In the discussion section, the authors comment on the sources of "background noise" in their data and ways to improve the specificity. They provide some analysis on this aspect in Supplementary figure S2. However, a better visualization of non-specificity in the sample could be a GO analysis of tissue-specificity, and presented as a pie chart as in Figure 2A. Non-neuronal proteins such as MYO-2 or MYO-3 repeatedly show up on the "TurboID trained" lists in several biological replicates (Tables S2 and S3). If a major fraction of the proteins after subtraction of control lists are non-specific, that increases the likelihood that the "hits" observed are by chance. This analysis should be presented in one of the main figures as it is essential for the reader to gauge the reliability of the experiment.
We agree with this assessment and thank Reviewer 3 for this constructive suggestion. In response, we have now incorporated a comprehensive tissue-specific analysis of the learning proteome in the revised manuscript. Using the single neuron RNA-Seq database CeNGEN, we identified the proportion of neuronal vs non-neuronal proteins from each biological replicate of mass spectrometry data. Specifically, we present Table 1 __on page 17 (which we originally intended to include in the manuscript, but inadvertently left out), which shows that 87-95% (i.e. a large majority) of proteins identified across replicates corresponded to genes detected in neurons, supporting that the TurboID enzyme was able to target the neuronal proteome as expected. __Table 1 is now described in the main text of the revised work on page 16.
In addition, we performed neuron-specific analyses using both the WormBase gene enrichment tool and the CeNGEN single-cell transcriptomic database, which we describe in detail on our response to Reviewer 1 Major Comment 2. To summarise, these analyses revealed enrichment of several neuron classes, including those previously implicated in associative learning (e.g., ASEL, AIB, RIS, AVK) as well as neurons not previously studied in this context (e.g., IL1, DA9, DVC) (summarised in Table S7). By examining expression overlap across neuron types, we identified shared and distinct profiles that suggest potential functional connectivity and candidate circuits underlying behavioural plasticity (Figure 4). Taken together, these data show that the proteins identified in our dataset are (1) neuronal and (2) expressed in neurons that are known to be required for learning. Methods are detailed on pages 50-51.
Other than the above, the authors have provided sufficient details in their experimental and analysis procedures. They have performed appropriate controls, and their data has sufficient biological and technical replaictes for statistical analysis.
We appreciate this positive feedback and thank the Reviewer for acknowledging the clarity of our experimental and analysis procedures.
Minor Comments:
There is an error in the first paragraph of the discussion, in the sentences discussing the learning effects in gar-1 mutant worms. The sentences in lines 12-16 on page 22 says that gar-1 mutants have improved salt-associative learning and defective salt-aversive learning, while in fact the data and figures state the opposite.
We appreciate the Reviewer noting this discrepancy. As clarified in our response to Reviewer 1, Major Comment 1 above, we reanalysed the behavioural data to ensure consistency across genotypes by comparing only those tested within the same biological replicates (thus having the same N for all genotypes). Upon this reanalysis, we found that the previously reported phenotype for gar-1 mutants in salt-associative learning was not statistically different from wild-type controls. Therefore, we have removed references to GAR-1 from the manuscript.
__Reviewer #3 (Significance (Required)): __Strengths and limitations: This study used neuron-specific TurboID expression with transient biotin exposure to capture a temporally restricted snapshot of the C. elegans nervous system proteome during salt-associative learning. This is an elegant method to identify proteins temporally specific to a certain condition. However, there are several limitations in the way the experiments and analyses were performed which affect the reliability of the data. As the authors themselves have noted in the discussion, background noise is a major issue and several steps could be taken to improve the noise at the experimental or analysis steps (use of integrated C. elegans lines to ensure uniformity of samples, flow cytometry to isolate neurons, quantitative mass spec to detect fold change vs. strict presence/absence). Advance: Several studies have demonstrated the use of proximity labeling to map the interactome by using a bait protein fusion. In fact, expressing TurboID not fused to a bait protein is often used as a negative control in proximity labeling experiments. However, this study demonstrates the use of free TurboID molecules to acquire a global snapshot of the proteome under a given condition. Audience: Even with the significant limitations, this study is specifically of interest to researchers interested in understanding learning and memory formation. Broadly, the methods used in this study could be modified to gain insights into the proteomic profiles at other transient developmental stages. The reviewer's field of expertise: Cell biology of C. elegans neurons.
We thank the reviewer for their thoughtful evaluation of our work. We appreciate the recognition of the novelty and potential of using neuron-specific TurboID to capture a temporally restricted snapshot of the C. elegans nervous system proteome during learning. We agree that this approach offers a unique opportunity to identify proteins associated with specific behavioural states in future studies.
We also appreciate the reviewer’s comments regarding limitations in experimental and analytical design. In revising the manuscript, we have taken several steps to address these concerns and improve the clarity, rigour, and interpretability of our data. Specifically:
Reviewer #4 (Evidence, reproducibility and clarity (Required)):
Summary:
In this manuscript, authors used a learning paradigm in C. elegans; when worms were fed in a saltless plate, its chemotaxis to salt is greatly reduced. To identify learning-related proteins, authors employed nervous system-specific transcriptome analysis to compare whole proteins in neurons between high-salt-fed animals and saltless-fed animals. Authors identified "learning-specific genes" which are observed only after saltless feeding. They categorized these proteins by GO analyses and pathway analyses, and further stepped forward to test mutants in selected genes identified by the proteome analysis. They find several mutants that are defective or hyper-proficient for learning, including acc-1/3 and lgc-46 acetylcholine receptors, gar-1 acetylcholine receptor GPCR, glna-3 glutaminase involved in glutamate biosynthesis, and kin-2, a cAMP pathway gene. These mutants were not previously reported to have abnormality in the learning paradigm.
Major comments:
1) There are problems in the data processing and presentation of the proteomics data in the current manuscript which deteriorates the utility of the data. First, as the authors discuss (page 24, lines 5-12), the current approach does not consider amount of the peptides. Authors state that their current approach is "conservative", because some of the proteins may be present in both control and learned samples but in different amounts. This reviewer has a concern in the opposite way: some of the identified proteins may be pseudo-positive artifacts caused by the analytical noise. The problem is that authors included peptides that are "present" in "TurboID, trained" sample but "absent" in the "Non-Tg, trained" and "TurboID, control" samples in any one of the biological replicates, to identify "learning proteome" (706 proteins, page 8, last line - page 9, line 8; page 32, line 21-22). The word "present" implies that they included even peptides whose amounts are just above the detection threshold, which is subject to random noise caused by the detector or during sample collection and preparation processes. This consideration is partly supported by the fact that only a small fraction of the proteins are common between biological replicates (honestly and respectably shown in Figure S2). Because of this problem, there is no statistical estimate of the identity in "learning proteome" in the current manuscript. Therefore, the presentation style in Tables S2 and S3 are not very useful for readers, especially because authors already subtracted proteins identified in Non-Tg samples, which must also suffer from stochastic noise. I suggest either quantifying the MS/MS signal, or if authors need to stick to the "present"/"absent" description of the MS/MS data, use the number of appearances in biological replicates of each protein as estimate of the quantity of each protein. For example, found in 2 replicates in "TurboID, learned" and in 0 replicates in "Non-Tg, trained". One can apply statistics to these counts. This said, I would like to stress that proteins related to acquisition of memory may be very rare, especially because learning-related changes likely occur in a small subset of neurons. Therefore, 1 time vs 0 time may be still important, as well as something like 5 times vs 1 time. In summary, quantitative description of the proteomics results is desired.
We thank the reviewer for these valuable comments and suggestions.
We acknowledge that quantitative proteomics would provide beneficial information; however, as also indicated by Reviewer 1 (in cross-comment), it is practically challenging to perform with TurboID. We have included discussion of potential future experiments involving quantitative mass spectrometry, as well as a comprehensive discussion of some of the limitations of our approach as summarised by this Reviewer, in the Discussion section (page 39). However, we note that our qualitative approach also provides beneficial knowledge, such as the identification of functional protein networks acting within biological pathways previously implicated in learning (Figure 2), and novel learning regulators ACC-1/3, LGC-46, and F46H5.3.
We agree with the assessment that the frequency of occurrence for each candidate we test per biological replicate is useful to disclose in the manuscript as a proxy for quantification. This was also highlighted by Reviewer 2 (Major Comment 1). As detailed above in response to R2, we have now separated candidates into two categories: ‘strong’ (present in 3 or more biological replicates) and ‘weak’ candidates (present in 2 or fewer biological replicates). We have also added behavioural data after testing 9 of these strong candidates in Figures 6 & S7.
We have also added Table 2 to the revised manuscript, which summarises the frequency-based representation of the proteomics results, as suggested. This is described on pages 22-23. Briefly, this shows the range of candidates further explored using single mutant testing. Specifically, this data showed that many of the tested candidates were more frequently detected in trained worms compared to high-salt controls. This includes both strong and weak candidates, providing a clearer view of how proteomic frequency informed our selection for functional testing.
2) There is another problem in the treatment of the behavioural data. In Experimental Procedures, authors state that they excluded data in which naive or control groups showed average CI 0.5499 for N2 (page 36, lines 5-7). How were these values determined? One common example for judging a data point as an outlier is > mean + 1.5, 2 or 3 SD, or Thank you for pointing this out. As mentioned by both Reviewer 1 and Reviewer 4, the original manuscript states the following: “Data was excluded for salt associative learning experiments when wild-type N2 displayed (1) an average CI ≤ 0.6499 for naïve or control groups and/or (2) an average CI either 0.5499 for trained groups.”
To clarify, we only excluded experiments in rare cases where N2 worms did not display robust high salt attraction before training, or where trained N2 did not display the expected behavioural difference compared to untrained or high-salt control N2. These anomalies were typically attributable to clear contamination or starvation issues that could clearly be observed prior to counting chemotaxis indices on CTX plates.
We established these exclusion criteria in advance of conducting multiple learning assays to ensure an objective threshold for identifying and excluding assays affected by these rare but observable issues. However, these criteria were later found to be unnecessary, as N2 worms robustly displayed the expected untrained and trained phenotypes for salt associative learning when not compromised by starvation or contamination.
We understand that the original criteria may have appeared to introduce arbitrary bias in data selection. To address this concern, we have removed these criteria from the revised manuscript from page 50.
Minor comments:
1) Related to Major comments 1), the successful effect of neuron-specific TurboID procedure was not evaluated. Authors obtained both TurboID and Non-Tg proteome data. Do they see enrichment of neuron-specific proteins? This can be easily tested, for example by using the list of neuron-specific genes by Kaletsky et al. (http://dx.doi.org/10.1038/nature16483 or http://dx.doi.org/10.1371/journal.pgen.1007559), or referring to the CenGEN data.
We thank this Reviewer for this helpful suggestion, which was echoed by Reviewer 3 (Major Comment 1). As indicated in the response to R3 above, the revised manuscript now includes Table 1 as a tissue-specific analysis of the learning proteome, using the single neuron RNA-Seq database CeNGEN to identify the proportion of neuronal proteins from each biological replicate of mass spectrometry data. Generally, we observed a range of 87-95% of proteins corresponded to genes from the CeNGEN database that had been detected in neurons, providing evidence that the TurboID enzyme was able to target the neuronal proteome as expected. Table 1 is now described in the main text of the revised work on pages 16 & 17.
2) The behavioural paradigm needs to be described accurately. Page 5, line 16-17, "C. elegans normally have a mild attraction towards higher salt concentration": in fact, C. elegans raised on NGM plates, which include approximately 50mM of NaCl, is attracted to around 50mM of NaCl (Kunitomo et al., Luo et al.) but not 100-200 mM.
We thank the Reviewer for pointing this out. We agree that clarification is necessary. The revised text reads as follows on page 5: “C. elegans are typically grown in the presence of salt (usually ~ 50 mM) and display an attraction toward this concentration when assayed for chemotaxis behaviour on a salt gradient (Kunitomo et al., 2013, Luo et al., 2014). Training/conditioning with ‘no salt + food’ partially attenuates this attraction (group referred to ‘trained’).”
Authors call this assay "salt associative learning", which refers to the fact that worms associate salt concentration (CS) and either presence or absence of food (appetitive or aversive US) during conditioning (Kunitomo et al., Luo et al., Nagashima et al.) but they are looking at only association with presence of food, and for proteome analysis they only change the CS (NaCl concentration, as discussed in Discussion, p24, lines 4-5). It is better to attempt to avoid confusion to the readers in general.
Thank you Reviewer 4 for highlighting this clarity issue. We clarify our definition of “salt associative learning” for the purpose of this study in the revised manuscript on page 6 with the following text:
“Similar behavioural paradigms involving pairings between salt/no salt and food/no food have been previously described in the literature (Nagashima et al. 2019). Here, learning experiments were performed by conditioning worms with either ‘no salt + food’ (referred to as ‘salt associative learning’) or ‘salt + no food’ (called ‘salt aversive learning’).”
3) page 32, line 23: the wording "excluding" is obscure and misleading because the elo-6 gene was included in the analysis.
We appreciate this Reviewer for pointing out this misleading comment, which was unintentional. We have now removed it from the text (on page 21).
4) Typo at page 24, line 18: "that ACC-1" -> "than ACC-1".
This has been corrected (on page 37).
5) Reference. In "LEO, T. H. T. et al.", given and sir names are flipped for all authors. Also, the paper has been formally published (http://dx.doi.org/10.1016/j.cub.2023.07.041).
We appreciate the Reviewer drawing our attention to this – the reference has been corrected and updated.
I would like to express my modest cross comments on the reviews:
1) Many of the reviewers comment on the shortage in the quantitative nature of the proteome analysis, so it seems to be a consensus.
Thank you Reviewer 4 for this feedback. We appreciate the benefit in performing quantitative mass spectrometry, in that it provides an additional way to parse molecular mechanisms in a biological process (e.g., fold-changes in protein expression induced by learning). However, we note that quantitative mass spectrometry is challenging to integrate with TurboID due to the requirement to enrich for biotinylated peptides during sample processing (we now mention this on page 39). Nevertheless, it would be exciting to see this approach performed in a future study.
To address the limitations of our original qualitative approach and enhance the clarity and utility of our dataset, we have made the following revisions in the manuscript:
2) Also, tissue- or cell-specificity of the identified proteins were commonly discussed. In reviewer #3's first Major comment, appearance of non-neuronal protein in the list was pointed out, which collaborate with my (#4 reviewer's) question on successful identification of neuronal proteins by this method. On the other hand, reviewer #1 pointed out subset neuron-specific proteins in the list. Obviously, these issues need to be systematically described by the authors.
We agree with Reviewer 4 that these analyses provide a critical angle of analysis that is not explored in the original manuscript.
Tissue analysis (Reviewer 3 Major Comment 1): We have used the single neuron RNA-Seq database CeNGEN, to identify that 87-95% (i.e. a large majority) of proteins identified across replicates corresponded to genes detected in neurons. These findings support that the TurboID enzyme was able to target the neuronal proteome as expected. Table 1 provides this information as is now described in the main text of the revised work on page 16.
__Neuron class analyses (Reviewer 1 Major Comment 2): __In response, we have used the suggested Wormbase gene enrichment tool and CeNGEN. We specifically input proteins from the learning proteome into Wormbase, after filtering for proteins unique to TurboID trained animals. For CeNGEN, we compared genes/proteins from control worms and trained worms to identify potential neurons that may be involved in this learning paradigm.
Briefly, we found highlight a range of neuron classes known in learning (e.g., RIS interneurons), cells that affect behaviour but have not been explored in learning (e.g., IL1 polymodal neurons), and neurons for which their function/s are unknown (e.g., pharyngeal neuron I3). Corresponding text for this new analysis has been added on pages 16-20, with a new table and figure added to illustrate these findings (Table S7 & Figure 4). Methods are detailed on pages 50-51.
3) Given reviewer #1's OPTIONAL Major comment, as an expert of behavioral assays in C. elegans, I would like to comment based on my experience that mutants received from Caenorhabditis Genetics Center or other labs often lose the phenotype after outcrossing by the wild type, indicating that a side mutation was responsible for the observed behavioral phenotype. Therefore, outcrossing may be helpful and easier than rescue experiments, though the latter are of course more accurate.
Thank you for this suggestion. To address the potential involvement of background mutations, we have done experiments with backcrossed versions of mutants tested where possible, as shown in Figure 6. We found that F46H5.3(-) mutants maintained enhanced learning capacity after backcrossing with wild type, compared to their non-backcrossed mutant line. This was in contrast to C30G12.6(-) animals which lost their enhanced learning phenotype following backcrossing using wild type worms. This is described in the text on pages 24-26.
4) Just let me clarify the first Minor comment by reviewer #2. Authors described that the kin-2 mutant has abnormality in "salt associative learning" and "salt aversive learning", according to authors' terminology. In this comment by reviewer #2, "gustatory associative learning" probably refers to both of these assays.
Reviewer 4 is correct. We have amended the wording appropriately on page 31 to clarify our meaning to address Reviewer 2’s comment.
5) There seem to be several typos in reviewer #1's Minor comments.
"In Page 9, Lines 17-18" -> "Page 8, Lines 17-18".
"Page 8, Line 24" -> "Page 7, Line 24".
"I would suggest to remove figure 3" -> "I would suggest to remove figure 2"
"summary figure similar to Figure 4" -> "summary figure similar to Figure 3"
"In the discussion Page 24, Line 14" -> "In the discussion Page 23, Line 14"
(I note that because a top page was inserted in the "merged" file but not in art file for review, there is a shift between authors' page numbers and pdf page numbers in the former.)
It would be nice if reviewer #1 can confirm on these because I might be wrong.
We appreciate Reviewer 4 noting this, and can confirm that these are the correct references (as indicated by Reviewer 1 in their cross-comments)
Reviewer #4 (Significance (Required)):
1) Total neural proteome analysis has not been conducted before for learning-induced changes, though transcriptome analysis has been performed for odor learning (Lakhina et al., http://dx.doi.org/10.1016/j.neuron.2014.12.029). This guarantees the novelty of this manuscript, because for some genes, protein levels may change even though mRNA levels remain the same. We note an example in which a proteome analysis utilizing TurboID, though not the comparison between trained/control, has led to finding of learning related proteins (Hiroki et al., http://dx.doi.org/10.1038/s41467-022-30279-7). As described in the Major comments 1) in the previous section, improvement of data presentation will be necessary to substantiate this novelty.
We appreciate this thoughtful feedback. We agree that while the neuronal transcriptome has been explored in Lakhina et al., 2015 for C. elegans in the context of memory, our study represents the first to examine learning-induced changes in the total neuronal proteome. We particularly agree with the statement that “for some genes, protein levels may change even though mRNA levels remain the same”. This is essential rationale that we now discuss on page 42.
Additionally, we acknowledge the relevance of the study by Hiroki et al., 2022, which used TurboID to identify learning-related proteins, though not in a trained versus control comparison. Our work builds on this by directly comparing trained and control conditions, thereby offering new insights into the proteomic landscape of learning. This is now clarified on page 36.
To substantiate the novelty and significance of our approach, we have revised the data presentation throughout the manuscript, including clearer candidate selection criteria, frequency-based representation of proteomic hits (Table 2), and neuron-specific enrichment analyses (Table S7 & Figure 4). We hope these improvements help convey the unique contribution of our study to the field.
2) Authors found six mutants that have abnormality in the salt learning (Fig. 4). These genes have not been described to have the abnormality, providing novel knowledge to the readers, especially those who work on C. elegans behavioural plasticity. Especially, involvement of acetylcholine neurotransmission has not been addressed. Although site of action (neurons involved) has not been tested in this manuscript, it will open the venue to further determine the way in which acetylcholine receptors, cAMP pathway etc. influences the learning process.
Thank you Reviewer 4, for this encouraging feedback. To further strengthen the study and expand its relevance, we have tested additional mutants in response to Reviewer 3’s comments, as shown in Figures 6 & S7. These results provide even more candidate genes and pathways for future exploration, enhancing the significance and impact of our study.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
In this manuscript, authors used a learning paradigm in C. elegans; when worms were fed in a saltless plate, its chemotaxis to salt is greatly reduced. To identify learning-related proteins, authors employed nervous system-specific transcriptome analysis to compare whole proteins in neurons between high-salt-fed animals and saltless-fed animals. Authors identified "learning-specific genes" which are observed only after saltless feeding. They categorized these proteins by GO analyses and pathway analyses, and further stepped forward to test mutants in selected genes identified by the proteome analysis. They find several mutants that are defective or hyper-proficient for learning, including acc-1/3 and lgc-46 acetylcholine receptors, gar-1 acetylcholine receptor GPCR, glna-3 glutaminase involved in glutamate biosynthesis, and kin-2, a cAMP pathway gene. These mutants were not previously reported to have abnormality in the learning paradigm.
Major comments:
Minor comments:
Cross-Commenting
I would like to express my modest cross comments on the reviews:
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
In the manuscript titled "Identifying regulators of associative learning using a protein-labelling approach in C. elegans" the authors attempted to generate a snapshot of the proteomic changes that happen in the C. elegans nervous system during learning and memory formation. They employed the TurboID-based protein labeling method to identify the proteins that are uniquely found in samples that underwent training to associate no-salt with food, and consequently exhibited lower attraction to high salt in a chemotaxis assay. Using this system they obtained a list of target proteins that included proteins represented in molecular pathways previously implicated in associative learning. The authors then further validated some of the hits from the assay by testing single gene mutants for effects on learning and memory formation.
Major comments:
In the discussion section, the authors comment on the sources of "background noise" in their data and ways to improve the specificity. They provide some analysis on this aspect in Supplementary figure S2. However, a better visualization of non-specificity in the sample could be a GO analysis of tissue-specificity, and presented as a pie chart as in Figure 2A. Non-neuronal proteins such as MYO-2 or MYO-3 repeatedly show up on the "TurboID trained" lists in several biological replicates (Tables S2 and S3). If a major fraction of the proteins after subtraction of control lists are non-specific, that increases the likelihood that the "hits" observed are by chance. This analysis should be presented in one of the main figures as it is essential for the reader to gauge the reliability of the experiment.
Other than the above, the authors have provided sufficient details in their experimental and analysis procedures. They have performed appropriate controls, and their data has sufficient biological and technical replaictes for statistical analysis.
Minor comments:
There is an error in the first paragraph of the discussion, in the sentences discussing the learning effects in gar-1 mutant worms. The sentences in lines 12-16 on page 22 says that gar-1 mutants have improved salt-associative learning and defective salt-aversive learning, while in fact the data and figures state the opposite.
Strengths and limitations:
This study used neuron-specific TurboID expression with transient biotin exposure to capture a temporally restricted snapshot of the C. elegans nervous system proteome during salt-associative learning. This is an elegant method to identify proteins temporally specific to a certain condition. However, there are several limitations in the way the experiments and analyses were performed which affect the reliability of the data. As the authors themselves have noted in the discussion, background noise is a major issue and several steps could be taken to improve the noise at the experimental or analysis steps (use of integrated C. elegans lines to ensure uniformity of samples, flow cytometry to isolate neurons, quantitative mass spec to detect fold change vs. strict presence/absence).
Advance:
Several studies have demonstrated the use of proximity labeling to map the interactome by using a bait protein fusion. In fact, expressing TurboID not fused to a bait protein is often used as a negative control in proximity labeling experiments. However, this study demonstrates the use of free TurboID molecules to acquire a global snapshot of the proteome under a given condition.
Audience:
Even with the significant limitations, this study is specifically of interest to researchers interested in understanding learning and memory formation. Broadly, the methods used in this study could be modified to gain insights into the proteomic profiles at other transient developmental stages.
The reviewer's field of expertise: Cell biology of C. elegans neurons.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
In this study by Rahmani in colleagues, the authors sought to define the "learning proteome" for a gustatory associative learning paradigm in C. elegans. Using a cytoplasmic TurboID expressed under the control of a pan-neuronal promoter, the authors labeled proteins during the training portion of the paradigm, followed by proteomics analysis. This approach revealed hundreds of proteins potentially involved in learning, which the authors describe using gene ontology and pathways analysis. The authors performed functional characterization of some of these genes for their requirement in learning using the same paradigm. They also compared the requirement for these genes across various learning paradigms, and found that most hits they characterized appear to be specifically required for the training paradigm used for generating the "learning proteome".
Major Comments:
Minor Comments:
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
Rahmani et al., utilize the TurboID method to characterize the global proteome changes in the worm's nervous system induced by a salt-based associative learning paradigm. Altogether, Rahmani et al., uncover 706 proteins that are tagged by the TurboID method specifically in samples extracted from worms that underwent the memory inducing protocol. Next, the authors conduct a gene enrichment analysis that implicates specific molecular pathways in salt-associative learning, such as MAP-kinase and cAMP-mediated pathways. The authors then screen a representative group of the hits from the proteome analysis. The authors find that mutants of candidate genes from the MAP-kinase pathway, namely dlk-1 and uev-3, do not affect the performance in the learning paradigm. Instead multiple acetylcholine signaling mutants significantly affected the performance in the associative memory assay, e.g., acc-1, acc-3, gar-1, and lgc-46. Finally, the authors demonstrate that the acetylcholine signaling mutants did not exhibit a phenotype in similar but different conditioning paradigms, such as aversive salt-conditioning or appetitive odor conditioning, suggesting their effect is specific to appetitive salt conditioning.
Major comments:
Minor comments:
1.Lack of clarity regarding the validation of the biotin tagging of the proteome. The authors show in Figure 1 that they validated that the combination of the transgene and biotin allows them to find more biotin-tagged proteins. However there is significant biotin background also in control samples as is common for this method. The authors mention they validated biotin tagging of all their experiments, but it was unclear in the text whether they validated it in comparison to no-biotin controls, and checked for the fold change difference. Also, it was unclear which exact samples were tested per replicate. In Page 9, Lines 17-18: "For all replicates, we determined that biotinylated proteins could be observed ...", But in Page 8, Line 24 : "We then isolated proteins from ... worms per group for both 'control' and 'trained' groups,... some of which were probed via western blotting to confirm the presence of biotinylated proteins". - Could the authors specify which samples were verified and clarify how? - OPTIONAL: include the fold changes of biotinylated proteins of all the ones that were tested. Similar to Figure 1.C. 2.Figure 2 does not add much to the reader, it can be summarized in the text, as the fraction of proteins enriched for specific cellular compartments. - I would suggest to remove figure 3 to text, or transfer it to the supplementry material. - OPTIONAL: I would suggest the authors to mark in a pathway summary figure similar to Figure 4 the results from the behavior assay of the genetic screen. This would allow the reader to better get the bigger picture and to connect to the systemic approach taken in Figures 2 and 3. 3. Typo in Figure 3: the circle of PPM1: The blue right circle half is bigger than the left one. 4. Unclarity in the discussions. In the discussion Page 24, Line 14, the authors raise this question: "why are the proteins we identified not general learning regulators?. The phrasing and logic of the argumentation of the possible answers was hard to follow. - Can you clarify?
Cross-Commenting
I would like to thank Reviewer #4 for the great cross comment summary, I find it accurate and helpful. I also would like to thank Reviewer #4 for spotting the typos in my minor comments, their page and figure numbers are the correct ones.
Small comment on common point 1 - My feeling is that it is challanging to do quantitative mass spectrometry, especially with TurboID. In general, the nature of MS data is that it hints towards a direction but a followup validation work is required in order to assess it. For example, I am not surprised that the fraction of repeats a hit appeared in does not predict well whether this hit would be validated behavioraly. Given these limitations, I find the authors' approach reasonable.
I also would like to highlight this major comment from reviewer 4: "In Experimental Procedures, authors state that they excluded data in which naive or control groups showed average CI < 0.6499, and/or trained groups showed average CI < -0.0499 or > 0.5499 for N2 (page 36, lines 5-7). " This threshold seems arbitrary to me too, and it requires the clarifications requested by reviewer 4.
This study does a great job to effectively utilize the TurboID technique to identify new pathways implicated in salt-associative learning in C. elegans. This technique was used in C. elegans before, but not in this context. The salt-associative memory induced proteome list is a valuable resource that will help future studies on associative memory in worms. Some of the implicated molecular pathways were found before to be involved in memory in worms like cAMP, as correctly referenced in the manuscript. The implication of the acetylcholine pathway is novel for C. elgeans, to the best of my knowledge. The finding that the uncovered genes are specifically required for salt associative memory and not for other memory assays is also interesting.
However overall I find the impact of this study limited. The premise of this work is to use the Turbo-ID method to conduct a systems analysis of the proteomic changes. The work starts by conducting network analysis and gene enrichment which fit a systemic approach. However, since the authors find that ~30% of the tested hits affect the phenotype, and since only 17/706 proteins were assessed, it is challenging to draw conclusive broad systemic claims. Alternatively, the authors could have focused on the positive hits, and understand them better, find the specific circuits where these genes act. This could have increased the impact of the work. Since neither of these two options are satisfied, I view this work as solid, but not wide in its impact and therefore estimate the audience of this study would be more specialized.
My expertise is in C. elegans behavior, genetics, and neuronal activity, programming and machine learning.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
The authors do not wish to provide a response at this time
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
In this study, Wasilewska and colleagues generated tmbim5-/- zebrafish line and demonstrated that tmbim5 loss of function leads to decrease in zebrafish size and induces muscle atrophy. Authors used immunohistochemistry to suggest that tmbim5-/- zebrafish shows reduced glycogen levels in muscle and liver. However, most of the immunohistochemistry is not quantitated and only qualitative differences are shown. Next, the authors measured mitochondrial calcium levels in the brain of tmbim5-/- zebrafish but there was no behavioral phenotype in the fish. It would have be better to measure mitochondrial calcium levels in the muscles of tmbim5-/- zebrafish as phenotype is muscle atrophy. Further, it is reported that the mitochondrial membrane potential and glycogen levels were perturbed in tmbim5-/- zebrafish.
Next, the authors generated a scl8b1-/- (a probable NCLX ortholog in zebrafish) zebrafish, which did not show any drastic phenotype. However, neither slc8b1 function nor the phenotype of scl8b1-/- zebrafish was well characterized. Further, authors created two double knockout zebrafish lines i.e. tmbim5-/-/mcu-/- and tmbim5-/-/slc8b1-/-. Interestingly, both these lines were viable and do not show any drastic phenotypes. The authors concluded that in these transgenic fishes compensatory and/or alternative mitochondrial Ca2+ mobilization pathways counterbalance the effects of silencing of these proteins.
Although it is an interesting study, the conclusions are not well supported with the data. At several places only qualitative images are shown and quantitative data is missing. Similarly, Ca2+ imaging in muscles of tmbim5-/- zebrafish is not performed. Finally, no molecular mechanism or molecular details are provided. Though Tmbim5's potential role in EMRE degradation is discussed, it is not experimentally investigated. The quality of the manuscript would significantly enhance if authors perform the suggested experiments.
Major Comments:
Minor Comments:
Referee cross-commenting
Several comments are common between the reviewers highlighting that those experiments are critical. Secondly, I agree with the concerns raised by other two reviewers.
In this study, authors report couple of new transgenic zebrafish lines. However, further characterization of slc8b1-/- is required. This study reinforces the existing idea that there are very robust compensatory mechanisms that maintain mitochondrial Ca2+ homeostasis. While the work provides useful insights, it could benefit from a broader scope to provide substantial advancement to existing knowledge.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary: The work of Wasilewska et al. focusses on the MCU independent basal Ca2+ uptake mechanisms and the effects of MCU, NCLX, and TMBIM5 KO on Zebrafish Ca2+ homeostasis, mortality, anatomy and metabolism. The authors found evidence that tmbim5 potentially has a bidirectional mode of operation and is able to extrude Ca2+ from the matrix as well as transfer Ca2+ into mitochondria. Further, a reduced membrane potential in tmbim5-/- fish and altered metabolism was found. While the conclusion drawn are well argumented, a few points have to be addressed.
Major Points:
Minor Points:
The authors claim that mRNA levels of mitochondrial proteins involved in Ca2+ transport in tmbim5-/- are unaffected (Figure EV3). While the T-tests show no significant alteration, what happens if a 2-way ANOVA shows a more general effect revealed between WT and TMBIM5-/-?
This is a well-designed and carefully executed piece of work. The experimental design is thoughtfully elaborated, and the topic is worthy of investigation. The strengths of this study lie in translating our knowledge of TMBIN5 from single cells to organism and organ function. Moreover, the work provides important new information that will help the scientific community working on mitochondrial regulation AND muscle diseases to understand how ions coordinately regulate mitochondrial function.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Although the experimental approach is promising (see below), the results do not significantly expand our current understanding. This is partly due to the challenges of interpreting negative results, which are nonetheless worth reporting. Some of the conclusions and interpretations of the results could benefit from further clarification and contextualization to enhance their impact:
The manuscript submitted by Wasilewska et al investigates the functional relationship between different mitochondrial calcium transporters using zebrafish as a model. The topic is of great interest. In the last 15 years, many mitochondrial calcium transporters have been identified. In some cases, their mechanism is not fully understood, such as in the case of TMBIM5, recently described by some as an H/Ca exchanger, or as a Ca channel by others. Furthermore, the functional relationship between different transporters has so far been studied in a partial and superficial way. I believe that this work is therefore of great interest because it aims to contribute to a fundamental problem that is still poorly studied. The idea of using zebrafish is interesting, as it is an organism that is easy to manipulate and phenotype, and because it is transparent, making it possible to use specific biosensors to characterize mitochondrial calcium dynamics, at least in principle. The paper therefore deserves attention.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
We thank the reviewers for their helpful comments and suggestions. Below you may find the point-by-point replies to their concerns.
Reviewer #1
“The research is meticulously conducted, and the data are compelling, as they demonstrate that the Nova-agrin-Lrp4-MuSK axis is also operational in non-vertebrates. The conclusions drawn by the authors are generally adequate; however, I find some instances of "it is the first time..." to be unnecessary.”
We have removed all unnecessary claims to that effect.
“The work also presents an unexpected finding that mouse Nova protein is unable to splice the Ciona agrin mini-gene (Figure 3). I believe the inability of mouse Nova1 and Nova2 to splice the Ciona agrin could also be due to insufficient expression levels of the mouse proteins. Therefore, the authors should include either a positive control (e.g., mouse agrin mini-gene) or demonstrate that the proteins are expressed at comparable levels.”
We have now included two additional datasets supporting our conclusion. First, we have included the positive control with the mouse Agrin minigene as suggested by the reviewer, which shows that mouse Nova1 and Nova2 are indeed still able to splice the mouse Agrin minigene in our assay (Figure 3C). Second, we included fluorescence images of the GFP-fused mouse Nova1 and Nova2 showing their proper expression in the cells (Figure S7).
“I am also not fully convinced that the model of autoinhibition for Ciona Nova is supported by sufficient experimental data. Again, there are no data showing that the levels of the various deletion mutants of Nova are consistent and hence, there could be issues with the stbsility of some of the deletion mutants and this could explain the observed difference in activity.”
We have added a few more datasets to further investigate the model. First, we have added an independent biological replicate of the “MLN” Nova isoform deletion mutant assays (Figure S8), as well as a separate assay using deletion mutants based on the “MMM” isoform (Figure S9). The results were consistent in both cases, confirming our initial observations. Next, we tested more directly the idea proposed by the reviewer that there are issues with stability, by looking at the fluorescence of the GFP-fused mutants. We did notice that the N/C-terminal deletion mutants were not expressed as well, but this was always mitigated by concurrent deletion of the KH3 domain. We have now expanded our discussion in the text to propose that there may be a negative effect of the KH3 domain on Nova expression/stability in the absence of the N/C termini. Although different from the model in which KH3 directly inhibits KH1/KH2, there does seem to be some inhibitory effect of KH3 on Nova expression/stability. “- In all schematic presentations, exon Z6 appears larger than exon Z5. However, Z6 is only 24 bp long, while Z5 is 3434 bp long. Please adjust this representation.”
To clarify, in Ciona Z6 is 18 bp long, and Z5 is 15 bp long, hence they code for 6 and 5 amino-acids, respectively. This is different from the mammalian Z exons, which may be the source of the confusion here. In our schematics, we are only representing the Ciona Z exons.
“- Is there consistency in the relative proportions of the 24-bp (Z6), 33-bp (Z5), and 57-bp (Z6 + Z5) PCR products? Studies in vertebrates have shown that AChR clustering activity is highest with the Z8 and Z19 products, while the Z11 product appears to be somewhat less active. It would be nice to also point out the different splice products are detected in Ciona.”
It was not clear if there was any consistency in the relative proportions of Ciona Agrin splice products in the minigene assays as performed in cultured mammalian cells, though in Figure 1 we have pointed out a more detailed characterization of the different splice products in vivo in Ciona. The different splice products’ confirmed sequences are also shown in the supplemental sequences file.
“Line 111: 'Z11' Agrin should be corrected to 'Z19' Agrin.”
To clarify again, we are only referring to the Ciona Agrin Z exons, which are not the same sizes as the mammalian Z exons. While Z19 would refer to the combination of exons Z8 and Z11 (8+11 = 19) in mammals, here in Ciona the equivalent combination is Z11 (Z5 + Z6).
“Line 168: "Figure H" should be updated to "Figure 2H."”
Fixed.
Reviewer #2:
__*“44 - ALS, references 8-12. These are old papers. A new review should be cited, either instead of in addition.”
*__
We have read some newer reviews and cite three more recent reviews (references 10-12) now.
__* 56 - "many" cases of CMS - some are not due to mutations in this pathway
*__
We have altered this to say “many”.
__* 57 - refs 29-46. This is a very large number of references for a point this is quite unimportant to the story. It would be better to cite recent reviews.
*__
We have removed some references and also cited more recent reviews here (references 38, 39).
__* 168 - should be 2H
*__
Fixed.
__* 205 - make N terminal extension more apparent in Figure 3D
*__
We have recolored the N terminus to be red, as to make it more apparent, in figures 3 and S8 and S9.
__* 235 - not a complete sentence
*__
Fixed.
308+ - can the authors clarify whether EBF knockdown has a selective effect on Nova vs general failure of the neurons to acquire a MN phenotype
We have been investigating this in a separate study on MN specification and differentiation in Ciona, which will be published as a preprint soon. EBF does not have a selective effect on Nova expression, as it appears to be regulating multiple aspects of neuronal differentiation, consistent with its role as previously studied in Ciona and other organisms (e.g. Kratsios et al. 2012, Catela et al. 2019, etc).__*
614 - explain in figure legend the decrease in apparent MR from left to right in 4B
*__
This is just an example of “bowed” or “curved” bands frequently seen in electrophoresis, usually due to uneven heat dissipation or other electrophoresis issues. However, the bands all correspond to the same products (Z+). We added an explanation in the legend.
General - three other key components of the pathway are MuSK, rapsyn, and DOK-7. Functional studies of these genes fall beyond the scope of this paper, but it would be helpful to know whether they are expressed in muscle and, if so, whether expression is muscle-specific.
We have added this to the discussion. While Musk and Dok-7 remain unstudied in Ciona, it has been shown that Rapsyn is muscle-specific in Ciona (Nishino et al. 2011).
Reviewer #3:
__*“1) The authors report two main Nova isoforms that seem to be produced by alternative promoters. They also claim that the MLN isoform is more strongly expressed in two of the studied conditions compared to the MMM protein (eggs and heart in Fig 1G), while both are equally abundant in st. 22.5 embryos and brain (Fig S1 and line 130). Therefore, both isoforms are likely involved in the regulation of the Agrin AS event. When performing the experiments that require to express the Nova protein, the authors choose to work with the "MLN" isoform arguing that it is more "ubiquitous" than the "MMM" isoform, although the last has a more evident nuclear localization signal (NLS) sequence. In the minigene analysis, the MLN isoform fails to produce transcripts with Z6 exon (which seems to be the most common Z+ isoform in the brain), and the amount of Z11-containing transcripts is very low compared to st. 22.5. Given that the N-terminal domain has a regulatory influence, as demonstrated by the authors, and that the MMM isoform is potentially more "neural-restricted" than the MLN, an intriguing possibility is that the MMM isoform might enhance the inclusion of Z6 and Z11 isoforms. To solve this issue, I suggest two experiments:
We have added additional minigene assay data using the MMM isoform (S9). We did not detect Z6 isoforms with MMM, though there may be slight differences in the ratio of Z5 and Z11 compared to the MLN assay. We believe this indicates that nuclear localization is not rate-limiting in our heterologous mammalian cell minigene assay, although it very well may regulate splicing activity more meaningfully in vivo in Ciona. This may be especially true in post-mitotic cells, as opposed to during embryogenesis when actively proliferating cells will break down and then reconstitute their nuclear envelopes over and over again, thus potentially allowing some of the MLN isoform to find its way into the nucleus. We still believe the production of the Z6 isoform may depend on additional Ciona-specific factors missing from the mammalian cells in our heterologous assay.
“- Test the regulatory activity of the upstream genomic region of exon 1a, in an equivalent way as for exon 1b in Fig 7A and B, to explore whether the promoter of the MMM isoform has a neural-restricted expression that could explain the AS pattern observed in st. 22.5 and brain.” We have done this, shown in Figure S15, which revealed that the promoter upstream of exon 1a (encoding the MMM isoform) drives only expression in mesenchyme and some epidermal cells, with no neuronal expression visible. This suggests that the majority of the neural expression is due to the cis-regulatory elements in the region between exons 1a and 1b. However, this region does not necessarily activate transcription only at exon 1b (encoding MLN isoform), as intronic elements can loop back and regulate transcription off “upstream” promoters. Thus we propose that the Nova [1b] -2011/+6 region drives expression of both MLN and MMM isoforms, though this remains to be fully tested. We believe the regulation and function of the different Nova isoforms in Ciona is beyond the scope of the current paper, though we are interested in investigating this more thoroughly in follow-up studies.
__*“2) The authors unveil the conservation of an Agrin AS event between mammals and a tunicate species with similar functional consequences for AChR clustering. While this is absolutely correct, the relatively low similarity of the AS exons between Ciona and mammals shown in Fig 1A may raise confusion or doubts in the readers regarding the homology of the event (as it did in my own case before I checked it in more detail). Therefore, an explicit alignment of both constitutive and alternative exons in a supplementary figure to clearly demonstrate the homology of the AS event across major taxonomic groups (with a few vertebrate and tunicate species) might help.
Furthermore, expression of Nova in motor neurons of amphioxus (Branchiostoma lanceolatum) was previously reported (ref. 60), and a quick look into publicly available Agrin transcripts (____https://www.ncbi.nlm.nih.gov/gene/136443694____) reveals a homologous AS event in this cephalochordate species.
C1 "Z7/Z6/Z8" C2 (partial)
Bla QADPAPLRQEGVG--LDGTTILNYPNAINK ... E-SNSIRE ... QEPNQDDNHFEVTFRTTSDHGLLLWNHKPGGG-DFIALAI Cro HSTDLLQDEQATAIYLDGTTKIMYRNAVKA ... --PNDFRE ... SRART-HNNYEIVFRTTARHGLLLMVGKAREGVDYIALAI Mmu IVEKSVGDLETLA--FDGRTYIEYLNAVTE ... ELTNEIPA ... EKALQ-SNHFELSLRTEATQGLVLWIGKVGERADYMALAI : . :** * : * **:. .*.: ... *::*: :** : :**:* * *::****
These two facts suggests a potential origin of the Nova-Agrin regulation at the base of the chordate phylum (and not restricted to Olfactores), which could be mentioned in the discussion as a relevant possibility.*__”
We thank the reviewer for this suggestion. Indeed, we have now added a more detailed alignment with Agrin sequences from more species in Figures S2 and S3, including amphioxus as so helpfully identified by the reviewer. We have added the observation that amphioxus Agrin appears to have a single Z exon encoding the NxI/V motif (no evidence for two Z exons as in tunicates or vertebrates). This indeed suggests that this pathway may be a chordate innovation, as we now discuss. We also add AlphaFold-assisted predictions of the NxF motif binding to the equivalent pocket in Lrp4 in both Ciona and mammals (Figure S1).
Line 168: Figure 2H instead of Figure H.
Fixed.
Line 287: "Taken together, these results reveal that a Nova-Agrin-Lrp4 pathway for AChR receptor clustering at the neuromuscular synapse is conserved from mammals to tunicates." While this sentence might be true, from mammals to tunicates might imply that it is conserved in all vertebrate and tunicate lineages, and this is not explored in the manuscript (there might be secondary losses). It would be more technically correct to say something similar like "...the neuromuscular synapse is conserved in the studied mammalian and tunicate lineages" or "...the neuromuscular synapse originated before the evolutionary divergence of tunicates and vertebrates"
We have fixed this now in a few places.
“Line 342. At the end of this paragraph, the possibility of conservation of the mechanism also in amphioxus could be discussed.”
We now discuss the amphioxus sequence and the idea that this mechanism was present in the last common chordate ancestor.
“Line 383: "the the apparent".”
Fixed.
“I agree that the mouse-specific agrin minigene to test the functionality of Nova1 and Nova2 would be a suitable positive control to discard protein stability/expression issues.”
We have tested this now with GFP fusion images (Figure S7) and using the mouse Agrin minigene (Figure 3C). Both indicate proper expression/splicing activity of mouse Nova1 and Nova2, supporting the idea that there is still some type of cross-species incompatibility as tested in mammalian cells.
“The only minor limitation, in my opinion, is that it lacks testing of the MMM Nova isoform in the minigene assay, to explore whether it has (or not) a complementary function to the MLN isoform that could fully explain the endogenous AS pattern.”
We have added MMM minigene assays, and these were largely identical to MLN assays. We propose that the N-terminus and nuclear localization do not significantly impact activity of Ciona Nova as tested in mammalian cells, however we cannot exclude the possibility that things may be different in vivo in Ciona.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary: The manuscript reports the conservation of the Nova-Agrin-Lrp4 pathway for AChR clustering in neuromuscular junctions beyond vertebrates, using the tunicate Ciona robusta as a model species. In addition, it also reveals Ebf as a key transcriptional activator of Nova in the motor neurons of Ciona. One of the main focuses of the work is the detailed study of an alternative splicing event in the Agrin gene of Ciona, demonstrating its regulation by Nova and the developmental cascade of consequences in AChR clustering due to misregulation of the Nova-Agrin-Lrp4 pathway through multiple functional experiments. Furthermore, it also explores molecular elements involved in the regulation of this event in trans and cis, including the KH and N-terminal domains of Nova (and their interactions) and the intronic YCAY binding domains.
Major comments: The claims and conclusions of the manuscript are generally very well supported with appropriate and reproducible functional experiments. For instance, the work demonstrates a key regulatory link between Nova and the studied AS event of Agrin using both a minigene system in human cells and a set of CRISPR/Cas9 Ciona mutants. Although analyzing mosaic embryos can be challenging, the authors successfully test different combinations of gRNAs to achieve efficient mutagenesis. Moreover, using the AChRA1::GFP clusters to measure the impact of the different mutants is very convincing.
While generally very robust and satisfying, the manuscript could benefit from addressing a few issues to improve its quality:
Furthermore, expression of Nova in motor neurons of amphioxus (Branchiostoma lanceolatum) was previously reported (ref. 60), and a quick look into publicly available Agrin transcripts (https://www.ncbi.nlm.nih.gov/gene/136443694) reveals a homologous AS event in this cephalochordate species.
C1 "Z7/Z6/Z8" C2 (partial)
Bla QADPAPLRQEGVG--LDGTTILNYPNAINK ... E-SNSIRE ... QEPNQDDNHFEVTFRTTSDHGLLLWNHKPGGG-DFIALAI Cro HSTDLLQDEQATAIYLDGTTKIMYRNAVKA ... --PNDFRE ... SRART-HNNYEIVFRTTARHGLLLMVGKAREGVDYIALAI Mmu IVEKSVGDLETLA--FDGRTYIEYLNAVTE ... ELTNEIPA ... EKALQ-SNHFELSLRTEATQGLVLWIGKVGERADYMALAI : . : * : * :. ..: ... ::: : : :: * ::***
These two facts suggests a potential origin of the Nova-Agrin regulation at the base of the chordate phylum (and not restricted to Olfactores), which could be mentioned in the discussion as a relevant possibility.
Minor comments:
Line 168: Figure 2H instead of Figure H.
Line 287: "Taken together, these results reveal that a Nova-Agrin-Lrp4 pathway for AChR receptor clustering at the neuromuscular synapse is conserved from mammals to tunicates." While this sentence might be true, from mammals to tunicates might imply that it is conserved in all vertebrate and tunicate lineages, and this is not explored in the manuscript (there might be secondary losses). It would be more technically correct to say something similar like "...the neuromuscular synapse is conserved in the studied mammalian and tunicate lineages" or "...the neuromuscular synapse originated before the evolutionary divergence of tunicates and vertebrates"
Line 342. At the end of this paragraph, the possibility of conservation of the mechanism also in amphioxus could be discussed.
Line 383: "the the apparent".
Referees cross-commenting
I agree that the mouse-specific agrin minigene to test the functionality of Nova1 and Nova2 would be a suitable positive control to discard protein stability/expression issues.
General assessment: This manuscript describes and demonstrates the deep evolutionary origin of a complex molecular pathway in the neuromuscular synapses of chordates. This work takes advantage of the broad genetic tools available in the tunicate Ciona robusta to support its main claims rigorously and strongly with a focused set of functional experiments. Moreover, it expands the known pathway revealing an upstream regulator of Nova in Ciona (Ebf) and opening a new research line in vertebrate motor neurons. The only minor limitation, in my opinion, is that it lacks testing of the MMM Nova isoform in the minigene assay, to explore whether it has (or not) a complementary function to the MLN isoform that could fully explain the endogenous AS pattern. Nevertheless, the current version of the manuscript is sufficiently robust to sustain its main conclusions.
Advance: Previous studies have reported deeply conserved AS events regulated by homologous tissue-specific splicing factors suggesting a putative similar function, such as the case of Esrp regulating the splicing of FGFRs in amphioxus and vertebrates (Burguera et al. 2017). However, to my knowledge, this work is the first to analyse the functional conservation of an alternative splicing event between chordate clades in its endogenous context while demonstrating an homologous ontogenetic role. In addition, it provides new insights of the molecular interactions between the N-terminal and KH domains of Nova and how they bind to the NISE elements in the Agrin pre-mRNA.
Audience: I consider this work interesting for a substantially broad audience, given that it reveals a surprisingly deep conservation of a molecular pathway across chordate lineages that is essential for the proper establishment of neuromuscular synapses. Thus, this study is imaginably interesting for evolutionary and molecular biologists, physiologists and even biomedical researchers that might be interested to explore a potential regulatory connection between Ebf genes and Nova in human motor neurons.
Fields of expertise: evo-devo, alternative splicing, chordates, transcriptomic and genome evolution.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary: Formation of the postsynaptic apparatus at the mammalian skeletal neuromuscular junction is controlled by a signaling pathway in which NOVA-mediated splicing generates an active form of the proteoglycan agrin (z-agrin), which is released from motoneurons, and interacts with LRP4 in muscle, leading to clustering of acetylcholine receptors beneath the nerve termina. This delightful paper demonstrates striking conservation of the pathway in the non-vertebrate chordate Ciona robusta, and also reveals some striking differences.
Major comments: none
Minor comments:
44 - ALS, references 8-12. These are old papers. A new review should be cited, either instead of in addition.
56 - "many" cases of CMS - some are not due to mutations in this pathway
57 - refs 29-46. This is a very large number of references for a point this is quite unimportant to the story. It would be better to cite recent reviews.
168 - should be 2H
205 - make N terminal extension more apparent in Figure 3D
235 - not a complete sentence
308+ - can the authors clarify whether EBF knockdown has a selective effect on Nova vs general failure of the neurons to acquire a MN phenotype
614 - explain in figure legend the decrease in apparent MR from left to right in 4B
General - three other key components of the pathway are MuSK, rapsyn, and DOK-7. Functional studies of these genes fall beyond the scope of this paper, but it would be helpful to know whether they are expressed in muscle and, if so, whether expression is muscle-specific.
The paper is complete, and the results are compelling, nicely explained, and carefully documented. My comments are minor.
It provides an interesting example of ways in which a pathway for synaptic development is conserved across distant vertebrate species, as well as ways inhich it is modified. I know of no other papers that do this.
Audience: basic research interested in neuroscience, evolution and development
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript examines the role of the splicing factor Nova in Ciona robusta, a tunicate that is the closest relative to vertebrates. The authors demonstrate the co-expression of Agrn and Nova mRNA during development in motor neurons, highlighting the correlative appearance of Nova-spliced exons Z6 and Z5. Importantly, CRISPR/Cas9-mediated inhibition of agrin splicing, deletion of its receptor Lrp4, and loss of Nova result in a significant reduction in the number of motor neuron-muscle synapses. This finding supports the notion that neuromuscular synapse formation is similarly regulated in non-vertebrates as it is in vertebrates. The authors subsequently investigate the domains within Nova responsible for agrin splicing and identify the transcription factor Ebf as a regulator of Nova expression in motor neurons.
The research is meticulously conducted, and the data are compelling, as they demonstrate that the Nova-agrin-Lrp4-MuSK axis is also operational in non-vertebrates. The conclusions drawn by the authors are generally adequate; however, I find some instances of "it is the first time..." to be unnecessary.
The work also presents an unexpected finding that mouse Nova protein is unable to splice the Ciona agrin mini-gene (Figure 3). I believe the inability of mouse Nova1 and Nova2 to splice the Ciona agrin could also be due to insufficient expression levels of the mouse proteins. Therefore, the authors should include either a positive control (e.g., mouse agrin mini-gene) or demonstrate that the proteins are expressed at comparable levels.
I am also not fully convinced that the model of autoinhibition for Ciona Nova is supported by sufficient experimental data. Again, there are no data showing that the levels of the various deletion mutants of Nova are consistent and hence, there could be issues with the stbsility of some of the deletion mutants and this could explain the observed difference in activity.
Minor Points:
Line 111: 'Z11' Agrin should be corrected to 'Z19' Agrin.
Line 168: "Figure H" should be updated to "Figure 2H."
Referees cross-commenting
In my view, the other reviews provide interesting insights that will further strengthen the manuscript.
This manuscript examines the role of the splicing factor Nova in Ciona robusta, a tunicate that is the closest relative to vertebrates. The authors demonstrate the co-expression of Agrn and Nova mRNA during development in motor neurons, highlighting the correlative appearance of Nova-spliced exons Z6 and Z5. Importantly, CRISPR/Cas9-mediated inhibition of agrin splicing, deletion of its receptor Lrp4, and loss of Nova result in a significant reduction in the number of motor neuron-muscle synapses. This finding supports the notion that neuromuscular synapse formation is similarly regulated in non-vertebrates as it is in vertebrates. The authors subsequently investigate the domains within Nova responsible for agrin splicing and identify the transcription factor Ebf as a regulator of Nova expression in motor neurons.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Note : The original preprint version of our manuscript has been reviewed by 3 subject experts for Review Commons. All the three reviewers’ comments on the original version of our manuscript have been fully addressed. Their input was extremely valuable in helping us clarify and refine the presentation of our results and conclusions. Their feedback contributed to making the study both more thoroughly developed and more accessible to a broad readership, while preserving its mechanistic depth. We believe that this revised version more effectively highlights the conceptual advances brought by our findings.
Reviewer #1
Evidence, reproducibility and clarity
The manuscript "Key roles of the zona pellucida and perivitelline space in promoting gamete fusion and fast block to polyspermy inferred from the choreography of spermatozoa in mice oocytes" by Dr. Gourier and colleagues explores the poorly understood process of gamete fusion and the subsequent block to polyspermy by live-cell imaging of mouse oocytes with intact zona pellucida in vitro. The new component in this study is the presence of the ZP, which in prior studies of live-cell imaging had been removed before. This allowed the authos to examine contributions of the ZP to the block in polyspermy in relation to the timing of sperm penetrating the ZP and sperm fusing with the oocyte. By carefully analysing the timing of the cascade of events, the authors find that the first sperm that reaches the membrane of the mouse oocyte is not necessarily the one that fertilizes the oocytes, revealing that other mechanisms post-ZP-penetration influence the success of individual sperm. While the rate of ZP penetration remains constant in unfertilized oocytes, it decreases upon fertilization for subsequent sperm, providing direct evidence for the known 'slow block to polyspermy' provided by changes to the ZP adhesion/ability to be penetrated. Careful statistical analyses allow the authors to revisit the role of the ZP in preventing polyspermy: They show that the ZP block resulting from the cortical reaction is too slow (in the range of an hour) to contribute to the immediate prevention of polyspermy in mice. The presented analyses reveal that the ZP does contribute to the block to polyspermy in two other ways, namely by effectively limiting the number of sperm that reach the oocyte surface in a fertilization-independent manner, and by retaining components like JUNO and CD9, that are shed from the oocyte plasma membrane after fertilization, in the perivitelline space, which may help neutralize surplus spermatozoa that are already present in the PVS. Lastly, the authors report that the ZP may also contribute to channeling the flagellar oscillations of spermatozoa in the PVS to promote their fusion competence.
Major comments:
The authors provide a careful analysis of the dynamics of events, though the analyses are correlative, and can only be suggestive of causation. While this is a limitation of the study, it provides important analysis for future research. Moreover, by analysing also control oocytes without fertilization and the timing of events, the authors have in some instances clear 'negative controls' for comparison.
Some claims would benefit from rewording or rephrasing to put the findings better in the context of what is already known and what is novel:
Only real-time imaging of in vitro fertilization of zona pellucida-intact oocytes, as performed in our study, is capable of determining which spermatozoon crossing the zona pellucida fuses with the oocyte. However, such studies are rare, and most do not specifically address this question. As Reviewers 1 & 3, we have not found any citation or reference telling or showing that it is not necessarily the first spermatozoon to penetrate the zona pellucida that fertilizes the egg. In contrast, at least one reference (Sato et al., 1979) explicitly reports the opposite. If, as suggested by Reviewer 1 and 3, it has indeed been observed before that the first sperm to pass the ZP is not always the one that fertilizes, and if this idea is generally accepted in the field, then it is all the more important that a study demonstrates and publishes this point. This is precisely what our study makes possible. However, in case we may have overlooked a previous reference making the same observation as ours, we have removed the phrasing ‘challenging prior dogma’. That being said, the key issue is not so much that it is not necessarily the first spermatozoon penetrating the perivitelline space that fertilizes, but rather why spermatozoa that successfully reach the PVS of an unfertilized oocyte may fail to achieve fertilization. This is one of the central questions our study sought to address.
We are concerned that we may disagree on this point. The penetration block resulting from cortical granule release progressively reduces the permeability of the zona pellucida to spermatozoa, relative to its baseline permeability prior to sperm–oocyte fusion. Any decrease in this baseline permeability occurring before the fusion block becomes fully effective can contribute to the prevention of polyspermy by limiting the number of sperm that can access the oolemma at a time when fusion is still possible. In contrast, once the fusion block is fully established, limiting the number of spermatozoa traversing the ZP becomes irrelevant regarding the block to polyspermy, as the fusion block alone is sufficient to prevent additional fertilizations, rendering the penetration block obsolete. The only scenario that could challenge this obsolescence is if the fusion block were transient. In that case, as Reviewer 1 suggests, the penetration block could indeed play a role at a later time-point. However, taken together, our study and that of Nozawa et al. (2018) support the conclusion that this is not the case in mice:
- Our in vitro study using kinetic tracking shows that the time constant for completion of the fusion block is typically 6.2 ± 1.3 minutes. During this time window, we observe that the permeability of the zona pellucida to spermatozoa does not yet decrease significantly from the baseline level it exhibited prior to sperm–oocyte fusion (see Figures 5B and S1B in the revised manuscript, and Figures 5A and 5B in the initial version). Consequently, before the fusion block is fully established, the penetration block can contribute only marginally—if at all—to the prevention of polyspermy. In contrast, the naturally low baseline permeability of the ZP—independent of any fertilization-triggered penetration block—as well as the relatively long timing of fusion ( minutes on average) after sperm penetration in the perivitelline space, are factors that contribute to the preservation of monospermic while the fusion block is still being established.
- Our in vitro study using kinetic tracking shows that once the fusion block is completed following the first fusion event, no additional spermatozoa are able to fuse with the oocyte until the end of the experiment, 4 hours post-insemination (see blue points and fitting curve in Figure 5C). Meanwhile, one or more additional spermatozoa—most of them motile and therefore viable—are present in the perivitelline space in 50% of the oocytes analyzed (purple point in Figure 5C). This demonstrates that, once established, the fusion block remains effective for at least the entire duration of the experiment, supporting the idea of a fully functional and long-lasting fusion block.
- Nozawa et al. (2018) found that female mice lacking ovastacin—the protease released during the cortical reaction that renders the zona pellucida impenetrable—are normally fertile. They additionally reported that the oocytes recovered from these females after mating are monospermic despite the systematic presence of additional spermatozoa in the perivitelline space. These findings further support the conclusion that in mice the fusion block is both permanent and sufficient to prevent polyspermy. For all these reasons, we believe that even at a later time-point, the penetration block does not contribute to the prevention of polyspermy in mice.
To clarify the fact that the penetration block does not necessarily contribute to prevent polyspermy, which indeed challenges the commonly accepted view, we have substantially revised the discussion. Furthermore, Figure 9 from the initial version of the manuscript has been replaced by Figure 8 in the revised version. This new figure provides a more didactic illustration of the inefficacy of the penetration block in preventing polyspermy in mice, by showing the respective impact of the fusion block, the penetration block, as well as fusion timing and the natural baseline permeability of the zona pellucida, on the occurrence of polyspermy.
As for the abstract, it has also been thoroughly revised. The content related to this section is now expressed in a way that emphasizes the factors that actively contribute to the prevention of polyspermy in mice, rather than those with no or marginal contribution (such as the penetration block in this case).
We thank reviewer 1 for pointing out the lack of precision in the abstract regarding the “components” released from the oolemma, and the fact that our phrasing may have given the impression that the post-fertilization release of CD9 and JUNO is a novel observation. The new observation is that CD9 and JUNO, which are known to be massively released from the oolemma after fertilization, bind to spermatozoa in the perivitelline space. However, we cannot rule out the possibility that other oocyte-derived molecules not investigated here may undergo a similar process. This is why we employed the broader term “components”, which encompasses both CD9 and JUNO as well as potential additional molecules. That said, we acknowledge the lack of precision introduced by this terminology. To address this, we have revised the corresponding sentence in the abstract to better reflect our new findings relative to previous ones, and to eliminate the ambiguity introduced by the word “component”.
The revised sentence of the abstract reads as follows:
“Our observation that non-fertilizing spermatozoa in the perivitelline space are coated with CD9 and JUNO oocyte’s proteins, which are known to be massively released from the oolemma after gamete fusion, supports the hypothesis that the fusion block involves an effective perivitelline space-block contribution consisting in the neutralization of supernumerary spermatozoa in the perivitelline space by these and potentially other oocyte-derived factors.”
Moreover, we cannot state in the abstract that the release of CD9 and JUNO occurs only after the fusion of the first spermatozoon and not before, since some CD9 and JUNO are already detectable in the perivitelline space (PVS) prior to fusion. What our study shows is that, before fertilization, CD9 and JUNO are predominantly localized at the oocyte membrane. In contrast, after fusion (four hours post-insemination), oocyte CD9 is distributed between the membrane and the PVS, and the only JUNO signal detectable in the oocyte is found in the PVS. This is what we describe in the Results section on page 15.
Regarding the acronym “OPM” in the initial version of the manuscript, although it was defined in the introduction as referring to the oocyte plasma membrane and not the outer plasma membrane (which, indeed, would not be meaningful), we acknowledge that it may have caused confusion to people in the field due to its resemblance to the commonly used meaningful acronym “OAM” for outer acrosomal membrane. To avoid any ambiguity, we have replaced the acronym “OPM” throughout the revised manuscript with the term “oolemma”, which unambiguously refers to the plasma membrane of the oocyte.
It is unclear to me what the relevance of dividing the post-fusion/post-engulfment into different phases as done in Fig 2 (phase 1, and phase 2) - also for the conclusions of this paper this seems rather irrelevant and overly complicated, since the authors never get back to it and don't need it (it's not related to the polyspermy block analyses). I would remove it from the main figures and not divide into those phases since it is distracting from the main focus.
Sperm engulfment and PB2 extrusion are two processes that follow sperm–oocyte fusion. As such, they are clear indicators that fusion has occurred and that meiosis has resumed. Their progression over time is readily identifiable in bright-field imaging: sperm engulfment is characterized by the gradual disappearance of the spermatozoon head from the oolemma, whereas PB2 extrusion is observed as the progressive emergence of a rounded protrusion from the oocyte membrane (Figure 2 in the initial manuscript and Figure S2 A&B in the revised version). The kinetics of these events, measured from the arrest of “push-up–like” movement of the sperm head against the oolemma —assumed to coincide with sperm-oocyte fusion, as further justified in a later response to Reviewer 1—provide reliable temporal landmarks for estimating the timing of fusion when the fusion event itself is not directly observed in real time (Figure S2 C&D).
The four landmarks used in this estimation are:
(i) the disappearance of the sperm head from the oolemma due to internalization (28 ± 2 minutes post-arrest, mean ± SD);
(ii) the onset of PB2 protrusion from the oolemma (28 ± 2 minutes post-arrest);
(iii) the moment when the contact angle between the PB2 protrusion and the oolemma shifts from greater than to less than 90° (49 ± 6 minutes post-arrest);
(iv) the completion of PB2 extrusion (73 ± 10 minutes post-arrest).
The approach used to determine the fusion time window of a fertilizing spermatozoon from these landmarks is detailed in the “Determination of the Fertilization Time Windows” section of the Materials and Methods. Compared to the initial version of the manuscript, we have added a paragraph explaining the rationale for using the arrest of the push-up–like movement as a reliable indicator for sperm–oocyte fusion and have clarified the description of the approach used to determine fertilization timing.
The timed characterization of sperm engulfment and PB2 extrusion kinetics is highly relevant to the analysis of the penetration and fusion blocks, however we agree that its place is more appropriate in the Supplementary Information than in the main text. In accordance with the reviewer’s recommendation, this section has therefore been moved to the Supplementary Information SI2.
For the statistical analysis, I am not sure whether the assumption "assumption that the probability distribution of penetration or fertilization is uniform within a given time window" is in fact true since the probability of fertilizing decreases after the first fertilization event.... Maybe I misunderstood this, but this needs to be explained (or clarified) better, or the limitation of this assumption needs to be highlighted.
During in vitro fertilization experiments with kinetic tracking, each oocyte is observed sequentially in turn. As a result, sperm penetration into the perivitelline space or fusion with the oolemma may occur either during an observation round or in the interval between two rounds. In the former case, penetration or fusion is directly observed in real time, allowing for high temporal precision in determining the moment of the event. In contrast, when penetration or fusion occurs between two observation rounds, the precise timing cannot be directly determined. We can only ascertain that the event took place within the time window we have determined. Because, within a given penetration or fusion time window, we do not know the exact moment at which the event occurred, there is no reason to favor one time over another. This justifies the assumption that all time points within the window are equally probable. This explanation has been added in the section Statistical treatment of penetration and fertilization chronograms to study the kinetics of fertilization, penetration block and fusion block of the main text and in the section Statistical treatment of penetrations and fertilizations chronograms to study penetration and fusion blocks of the material and methods.
-Suggestion for additional experiments:
If I understood correctly, the onset of fusion in Fig 2C is defined by stopping of sperm beating? If it is by the sudden stop of the beating flagellum, this should be confirmed in this situation (with the ZP intact) that it correctly defines the time-point of fusion since this has not been measured in this set-up before as far as I understand. In order to measure this accurately, the authors will need to measure this accurate to be able to acquire those numbers (of time from fusion to end of engulfment), e.g. by pre-loading the oocyte with Hoechst to transfer Hoechst to the fusing sperm upon membrane fusion.
The nuclear dye Hoechst is widely used as a marker of gamete fusion, as it transfers from the ooplasm—when preloaded with the dye—into the sperm nucleus upon membrane fusion, thereby signaling the happening of the fusion event. This technique is applicable in the context of in vitro fertilization using ZP-free oocytes. However, it is not suitable when cumulus–oocyte complexes are inseminated, as is the case in both in vitro experimental conditions of the present study (standard IVF and IVF with kinetic tracking). Indeed, when cumulus–oocyte complexes are incubated with Hoechst to preload the oocytes, the numerous surrounding cumulus cells also take up the dye. Consequently, upon insemination, spermatozoa acquire fluorescence while traversing and dispersing the cumulus mass—before reaching the ZP—thus rendering Hoechst labeling ineffective as a specific marker of membrane fusion. This remains true even under optimized conditions involving brief Hoechst incubation of cumulus–oocyte complexes ( Nonetheless, we have strong evidence supporting the use of the arrest of sperm movement as a surrogate marker for the moment of fusion. In our previous study (Ravaux et al., 2016; ref. 4 in the revised manuscript), we investigated the temporal relationship between the abrupt cessation of sperm head movement on the oolemma—resulting from strong flagellar beating arrest—and the fusion event, using ZP-free oocytes preloaded with Hoechst. That study revealed a temporal delay of less than one minute between the cessation of sperm oscillations and the actual membrane fusion, thereby supporting the conclusion that in ZP-free oocytes, the arrest of vigorous sperm movement at the oolemma is a reliable indicator of the moment at which fusion occurs. In the same study, the kinetics of sperm head internalization into the ooplasm were also characterized, typically concluding within 20–30 minutes after movement cessation. These findings are fully consistent with our current observations in ZP-intact oocytes, where sperm head engulfment was completed approximately 24 ± 3 minutes after the arrest of sperm oscillations. Taken together, these results strongly support the conclusion that, in both ZP-free and ZP-intact oocytes, the arrest of sperm movement is a reliable indicator of the fusion event. This assumption formed the basis for our determination of fertilization time points in the present study.
These justifications were not fully detailed in the original version of the manuscript. We have addressed this in the revised version by explicitly presenting this rationale in the Materials and Methods section under Determination of the Fertilization Time Windows.
Fig 8: 2 comments
We have followed this recommendation. Figure 8 of the initial manuscript has been replaced by Figure 6 in the revised manuscript, which illustrates the four situations encountered in this study: fertilized and unfertilized oocytes, each with and without unfused spermatozoa in their PVS. To better show JUNO/CD9 pre-fusion presence to the oocyte plasma membrane, as well as their post-fusion partial (for CD9) and near-complete (for JUNO) loss from the oocyte membrane (but persistence in the PVS), paired images of the same oocyte before and after of ZP removal are now provided, both for unfertilized (Figure 6A) and fertilized oocytes (Figure 6C).
As staining and confocal imaging of the oocytes were performed 4 hours after insemination, images of sperm in the PVS of an oocyte “pre-fertilization” cannot be strictly obtained. However, we can have images of spermatozoa present in the PVS of oocytes that remained unfertilized. This situation, now illustrated in Figure 6B of the revised manuscript, shows that these spermatozoa are also covered in JUNO and CD9, which they may have progressively acquired over time from the baseline presence of these proteins in the PVS of unfertilized oocytes. This also may provide a mechanistic explanation for their inability to fuse with the oolemma, and, consequently, for the failure of fertilization in these oocytes.
Minor comments:
We have followed this recommendation. The videos have been cropped and annotated in order to highlight the key events that support the points made in the result section from page 9 to 11 in the revised manuscript.
A general scheme addressing Reviewer 1 request, summarizing the key components and concepts discussed in the article and intended to help guide the reader, has been added to the introduction of the revised manuscript as Figure 1.
The title of the first Results section has been revised in accordance with Reviewer 1 suggestion. It now reads: Comparative study of penetration and fertilization rates under in vivo and two distinct in vitro fertilization conditions.
In the revised version of our manuscript, we have restructured this part of the analysis to ensure that more technical or secondary elements do not disrupt the flow of the main text. Accordingly, the equations have been reduced to only what is strictly necessary to understand our approach, their notation has been greatly simplified, and the statistical analysis of unfertilized oocytes whose zona pellucida was traversed by one or more spermatozoa has been moved to the Supplementary Information (SI1).
We agree with Reviewer 1 suggestion. Accordingly, we have not only thoroughly revised our abstract, but also the introduction and discussion, in order to better highlight the rationale of our study, its storyline, and the new findings which not only challenge certain established views but also open new research directions in the mechanisms of gamete fusion and polyspermy prevention.
Falls down has been removed from the new version and replaced with decreases
Significance
Overall, this manuscript provides very interesting and carefully obtained data which provides important new insights particularly for reproductive biology. I applaud the authors on first establishing the in vivo conditions (how often do multiple sperm even penetrate the ZP in vivo) since studies have usually just started with in vitro condition where sperm at much higher concentration is added to isolated oocyte complexes. Thank you for providing an in vivo benchmark for the frequency of multiple sperm being in the PVS. While this frequency is rather low (somewhat expectedly, with 16% showing 2-3 sperm in the PVS), this condition clearly exists, providing a clear rationale for the investigation of mechanisms that can prevent additional sperm from entering.
My own expertise is experimentally - thus I don't have sufficient expertise to evaluate the statistical methods employed here.
__ __
Reviewer #2
Evidence, reproducibility and clarity
Overall, this is a very interesting and relevant work for the field of fertilization. In general, the experimental strategies are adequate and well carried out. I have some questions and suggestions that should be considered before the work is published.
1) Why are the cumulus cells not mentioned when the AR is triggered before or while the sperms cross it? It seems the paper assumes from previous work that all sperm that reach ZP and the OPM have carried out the acrosome reaction. This, though probably correct, is still a matter of controversy and should be discussed. It is in a way strange that the authors do not make some controls using sperm from mice expressing GFP in the acrosome, as they have used in their previous work.
We do not mention the cumulus cells or whether the acrosome reaction is triggered before, during, or after their traversal (i.e., upon sperm binding to the ZP), as this question, while scientifically relevant, pertains to a distinct line of investigation that lies beyond the scope of the present study. Even with the use of spermatozoa expressing GFP in the acrosome, addressing this question would require a complete redesign of our kinetic tracking protocol, which was specifically conceived to monitor in bright field the dynamic behavior of spermatozoa from the moment they begin to penetrate the perivitelline space of an oocyte. Accordingly, we imaged oocytes that were isolated 15 minutes after insemination of the cumulus–oocyte complexes, by which time most (if not all) cumulus cells had detached from the oocytes, as explained in the fourth paragraph of the material and methods of both the initial and revised versions of the manuscript. The spermatozoa we had access to were therefore already bound to the zona pellucida at the time of removal from the insemination medium, and had thus necessarily passed through the cumulus layer. It is unclear for us why Reviewer 2 believes that we “assume from previous work that all sperm that reach ZP has carried out the acrosome reaction”. We could not find any statement in our manuscript suggesting, let alone asserting, such an assumption, which we know to be incorrect. Based on both published work from Hirohashi’s group in 2011 (Jin et al., 2011, DOI: 10.1073/pnas.1018202108) and our own unpublished observation (both involving cumulus-oocyte masses inseminated with spermatozoa expressing GFP in the acrosome), it is established that only a subset of spermatozoa reaching the ZP after crossing the cumulus layer has undergone acrosome reaction. Moreover, from the same sources—as well as from a recent publication by Buffone’s group (Jabloñsky et al., 2023 DOI: 10.7554/eLife.93792 ) which is the one to which reviewer 2 refers in her/his 3rd comment, it is also well established that spermatozoa have all undergone acrosome reaction when they enter the PVS. To the best of our knowledge, this latter point has long been widely accepted and is not questioned. Therefore, stating this in the first paragraph of the Discussion in the revised manuscript, while referencing the two aforementioned published studies, should be appropriate. What remains a matter of ongoing debate, however, is the timing and the physiological trigger(s) of the acrosome reaction in fertilizing spermatozoa. The 2011 study by Hirohashi’s group challenged the previously accepted view that ZP binding induces the acrosome reaction, showing instead that most spermatozoa capable of crossing the ZP and fertilizing the oocyte had already undergone the acrosome reaction prior to ZP binding. However, as this issue lies beyond the scope of our study, we do not consider it appropriate to include a discussion of it in the manuscript.
2) In the penetration block equations, it is not clear to me why (𝑡𝑃𝐹1) refers to both PIPF1 and 𝜎𝜎𝑃I𝑃𝐹1. Is it as function off?
That is correct: (tPF1) means function of the time post-first fertilization. Both the post-first fertilization penetration index (i.e. PIPF1) and its incertainty (i.e. 𝜎𝑃I𝑃𝐹1 ) vary as a function of this time. However, as mentioned in a previous response to Reviewer 1, this section has been rewritten to improve clarity and readability. The equations have been limited to those strictly necessary for understanding our approach, and their notation has been significantly simplified.
3) Why do the authors think that the flagella stops. The submission date was 2024-10-01 07:27:26 and there has been a paper in biorxiv for a while that merits mention and discussion in this work (bioRxiv [Preprint]. 2024 Jul 2:2023.06.22.546073. doi: 10.1101/2023.06.22.546073.PMID: 37904966).
Our experimental approach allows us to determine when the spermatozoon stops moving, but not why it stops. We thank Reviewer 3 for pointing out this very relevant paper from Buffone’s group (doi: 10.7554/eLife.93792) which shows the existence of two distinct populations of live, acrosome-reacted spermatozoa. These correspond to two successive stages, which occur either immediately upon acrosome reaction in a subset of spermatozoa, or after a variable delay in others, during which the sperm transitions from a motile to an immotile state. The transition from the first to the second stage was shown to follow a defined sequence: an increase in the sperm calcium concentration, followed by midpiece contraction associated with a local reorganization of the helical actin cortex, and ultimately the arrest of sperm motility. For fertilizing spermatozoa in the PVS, this transition was shown to occur upon fusion. However, it was also reported in some non-fertilizing spermatozoa that this transition took place within the PVS. These findings are consistent with the requirement for sperm motility in order to achieve fusion with the oolemma. Moreover, the fact that some spermatozoa may prematurely transition to the immotile state within the PVS can therefore be added to the list of possible reasons why a spermatozoon that penetrates the PVS of an oocyte might fail to fuse.
This discussion has been added to the first paragraph of the Discussion section of our revised manuscript.
4) Please correct at the beginning of Materials and Methos: Sperm was obtained from WT male mice, it should say were.
Thank you, the correction has been done.
5) This is also the case in the fourth paragraph of this section: oocyte were not was.
The sentence in question has been modified as followed: “In the in vitro fertilization experiments with kinetic tracking, a subset of oocytes—together with their associated ZP-bound spermatozoa—was isolated 15 minutes post-insemination and transferred individually into microdrops of fertilization medium to enable identification.”
Significance
Understanding mammalian gamete fusion and polyspermy inhibition has not been fully achieved. The authors examined real time brightfield and confocal images of inseminated ZP-intact mouse oocytes and used statistical analyses to accurately determine the dynamics of the events that lead to fusion and involve polyspermy prevention under conditions as physiological as possible. Their kinetic observations in mice gamete interactions challenge present paradigms, as they document that the first sperm is not necessarily the one that fertilizes, suggesting the existence of other post-penetration fertilization factors. The authors find that the zona pellucida (ZP) block triggered by the cortical reaction is too slow to prevent polyspermy in this species. In contrast, their findings indicate that ZP directly contributes to the polyspermy block operating as a naturally effective entry barrier inhibiting the exit from the perivitelline space (PVS) of components released from the oocyte plasma membrane (OPM), neutralizing unwanted sperm fusion, aside from any block caused by fertilization. Furthermore, the authors unveil a new important ZP role regulating flagellar beat in fertilization by promoting sperm fusion in the PVS.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
SUMMARY: This study by Dubois et al. utilizes live-cell imaging studies of mouse oocytes undergoing fertilization. A strength of this study is their use of three different conditions for analyses of events of fertilization: (1) eggs undergoing fertilization retrieved from females at 15 hr after mating (n = 211 oocytes); (2) cumulus-oocyte complexes inseminated in vitro (n = 220 oocytes), and (3) zona pellucida (ZP)-intact eggs inseminated in vitro, transferred from insemination culture once sperm were observed bound to the ZP for subsequent live-cell imaging (93 oocytes). This dataset and these analyses are valuable for the field of fertilization biology. Limitations of this manuscript are challenges arise with some conclusions, and the presentation of the manuscript. There are some factual errors, and also some places where clearer explanations should to be provided, in the text and potentially augmented with illustrations to provide more clarity on the models that the authors interpret from their data.
MAJOR COMMENTS:
The authors are congratulated on their impressive collection of data from live-cell imaging. However, the writing in several sections is challenging to understand or seems to be of questionable accuracy. The lack of accuracy is suspected to be more an effect of overly ambitious attempts with writing style, rather than to mislead readers. Nevertheless, these aspects of the writing should be corrected. There also are multiple places where the manuscript contradicts itself. These contradictions should be corrected. Finally, there are factual points from previous studies that need correction.
Second, certain claims and the conclusions as presented are not always clearly supported by the data. This may be connected to the issues with writing style, word and phrasing choices, etc. The conclusions could be expressed more clearly, and thus may not require additional experiments or analyses to support them. The authors might also consider illustrations as ways to highlight the points they wish to make. (Figure 7 is a strong example of how they use illustrations to complement the text).
In response to Reviewer 3's concern about the writing style, which made several sections difficult to understand, we have thoroughly revised the entire manuscript to improve clarity, and precision. To further enhance comprehension, we have added illustrations in the revised version of the manuscript:
Figure 1A presents the gamete components; Figure 1B depicts the main steps of fertilization considered in the present study; and Figure 1C illustrates the penetration and fusion blocks, along with the respective contributing mechanisms: the ZP-block for the penetration block, and the membrane-block and PVS-block for the fusion block
Figure 2A provides a description of the three experimental protocols used in this study: Condition 1, in vivo fertilization after mating; Condition 2, standard in vitro fertilization following insemination of cumulus-oocyte complexes; and Condition 3, in vitro fertilization with kinetic tracking of oocytes isolated from the insemination medium 15 min after insemination of the cumulus-oocyte complexes.
Figure 4 (formerly Figure 7 in the initial version) now highlights all fusing and non-fusing situations documented in videos 1-6 and associated paragraphs of the Results section.
In the Discussion, Figure 9 from the original version has been replaced by Figure 8, which now provides a more pedagogical illustration of the inefficacy of the penetration block in preventing polyspermy in mice. This figure illustrates the respective contributions of the fusion block, the penetration block, fusion timing, and the intrinsic permeability of the zona pellucida to the occurrence of polyspermy.
We hope that this revised version of the article will guide the reader smoothly throughout, without causing confusion.
Regarding the various points that Reviewer 3 perceives as contradictions or factual errors, or the claims and the conclusions which, as presented, should not always supported by the data, we will provide our perspective on each of them as they are raised in the review.
SPECIFIC COMMENTS:
(1) The authors should use greater care in describing the blocks to polyspermy, particularly because they appear to be wishing to reframe views about prevention of polyspermic fertilization. The title mentions of "the fast block to polyspermy;" this problematic for a couple of different reasons. There is no strong evidence for block to polyspermy in mammals that occurs quickly, particularly not in the same time scale as the first-characterized fast block to polyspermy. To many biologists, the term "fast block to polyspermy" refers to the block that has been described in species like sea urchins and frogs, meaning a rapid depolarization of the egg plasma membrane. However, such depolarization events of the egg membrane have not been detected in multiple mammalian species. Moreover, the change in the egg membrane after fertilization does not occur in as fast a time scale as the membrane block in sea urchins and frogs (i.e., is not "fast" per se), and instead occurs in a comparable time frame as the conversation of the ZP associated with the cleavage of ZP2. Thus, it is misleading to use the terms "fast block" and "slow block" when talking about mammalian fertilization. This also is an instance of where the authors contradict themselves in the manuscript, stating, "the membrane block and the ZP block are established in approximatively the same time frame" (third paragraph of Introduction). This statement is indeed accurate, unlike the reference to a fast block to polyspermy in mammals.
We fully agree with Reviewer 3 on the importance of clearly defining the two blocks examined in the present study—the penetration block and the fusion block (as referred to in the revised version) —and of situating them in relation to the three blocks described in the literature: the ZP-block, membrane-block, and PVS-block. We acknowledge that this distinction was not sufficiently clear in the original version of the manuscript. In the revised version, these two blocks and their relationship to the ZP-, membrane-, and PVS-blocks are now clearly introduced in the second paragraph of the Introduction section and illustrated in the first figure of the manuscript (Fig. 1C). They are then discussed in detail in two dedicated paragraphs of the Discussion, entitled Relation between the penetration block and the ZP-block and Relation between the fusion block and the membrane- and PVS-blocks.
The penetration block refers to the time-dependent decrease in the number of spermatozoa penetrating the perivitelline space (PVS) following fertilization, whereas the fusion block refers to the time-dependent decrease in sperm-oolemma fusion events after fertilization. It is precisely to the characterization of these two blocks that our in vitro fertilization experiments with kinetic tracking allow us to access.
In this study, as in the literature, fusion-triggered modifications of the ZP that hinder sperm traversal of the ZP are referred to as the ZP-block (also known as ZP hardening). The ZP-block thus contributes to the post-fertilization reduction in sperm penetration into the PVS and thereby underlies the penetration block. Similarly, fusion-triggered alterations of the PVS and the oolemma that reduce the likelihood of spermatozoa that have reached the PVS successfully to fuse with the oolemma are referred to as the PVS-block and membrane-block, respectively. These two blocks act together to reduce the probability of sperm-oolemma fusion after fertilization, and thus contribute to the fusion block.
The time constant of the penetration block was found to be 48.3 ± 9.7 minutes, which is consistent with the typical timeframe of ZP-block completion—approximately one hour post-fertilization in mice—as reported in the literature. By contrast, the time constant of the fusion block was determined to be 6.2 ± 1.3 minutes, which is markedly faster than the time typically reported in the literature for the completion of the fusion-block (more than one hour in mice). This strongly suggests that the kinetics of the fusion block are not primarily governed by its membrane-block component, but rather by its PVS-block component—about which little to nothing was previously known.
Contrary to what Reviewer 3 appears to have understood from our initial formulation, there is therefore no contradiction or error in stating that "the membrane block and the ZP block are established within approximately the same timeframe", while the fusion block, which proceeds much more rapidly, is likely to rely predominantly on the PVS-block. We have thoroughly revised the manuscript to clarify this key message of the study.
However, we understand Reviewer 3’s objection to referring to the fusion block (or the PVS-block) as a fast block, given that this term is conventionally reserved for the immediate fertilization-triggered membrane depolarization occurring in sea urchins and frogs. Although the kinetics we report for the fusion block are considerably faster than those of the penetration block, they occur on the scale of minutes, and not seconds. In line with the reviewer's recommendation, we have therefore modified both the title and the relevant passages in the text to remove all references to the term fast block in the revised version.
(2) The authors aim to make the case that events occurring in the perivitelline space (PVS) prevent polyspermic fertilization, but the data that they present is not strong enough to make this conclusion. Additional experiments would optional for this study, but data from such additional experiments are needed to support the authors' claims regarding these functions in fertilization. Without additional data, the authors need to be much more conservative in interpretations of their data. The authors have indeed observed phenomena (the presence of CD9 and JUNO in the PVS) that could be consistent with a molecular basis of a means to prevent fertilization by a second sperm. However, the authors would need additional data from additional experimental studies, such as interfering with the release of CD9 and JUNO and showing that this experimental manipulation leads to increased polyspermy, or creating an experimental situation that mimics the presence of CD9 and JUNO (in essence, what the authors call "sperm inhibiting medium" on page 20) and showing that this prevents fertilization.
A major section of the Results section here (starting with "The consequence is that ... ") is speculation. Rather than be in the Results section, this should be in the Discussion. The language should be also softened regarding the roles of these proteins in the perivitelline space in other portions of the manuscript, such as the abstract and the introduction.
Finally, the authors should do more to discuss their results with the results of Miyado et al. (2008), which interestingly, posited that CD9 is released from the oocytes and that this facilitates fertilization by rendering sperm more fusion-competent. There admittedly are two reports that present data that suggest lack of detection of CD9-containing exosomes from eggs (as proposed by Miyado et al.), but nevertheless, the authors should put their results in context with previous findings.
We generally agree with all the remarks and suggestions made here. In the revised version of the manuscript, we have retained in the Results section (pp. 14–15) only the factual data concerning the localization of CD9 and JUNO in unfertilized and fertilized oocytes, as well as in the spermatozoa present in the PVS of these oocytes. We have taken care not to include any interpretive elements in this section, which are now presented exclusively in a dedicated paragraph of the Discussion, entitled “Possible molecular bases of the membrane-block and ZP-block contributing to the fusion block” (p. 21). There, we develop our hypothesis and discuss it in light of both the findings from the present study and previous work by other groups. In doing so, we also address the data reported by Miyado et al. (2008, https://doi.org/10.1073/pnas.0710608105), as well as subsequent studies by two other groups—Gupta et al. (2009, https://doi.org/10.1002/mrd.21040) and Barraud-Lange et al. (2012, https://doi.org/10.1530/REP-12-0040)—that have challenged Miyado’s findings.
We are fully aware that our interpretation of the coverage of unfused sperm heads in the perivitelline space (PVS) by CD9 and JUNO, released from the oolemma—as a potential mechanism of sperm neutralization contributing to the PVS block—remains, at this stage, a plausible hypothesis or working model that, as such, warrants further experimental investigation. It is precisely in this spirit that we present it—first in the abstract (p.1), then in the Discussion section (p. 21), and subsequently in the perspective part of the Conclusion section (p. 22).
(3) Many of the authors' conclusions focus on their prior analyses of sperm interaction - beautifully illustrated in Figure 7. However, the authors need to be cautious in their interpretations of these data and generalizing them to mammalian fertilization as a whole, because mouse and other rodent sperm have sperm head morphology that is quite different from most other mammalian species.
In a similar vein, the authors should be cautious in their interpretations regarding the extension of these results to mammalian species other than mouse, given data on numbers of perivitelline sperm (ranging from 100s in some species to virtually none in other species), suggesting that different species rely on different egg-based blocks to polyspermy to varying extents. While these observations of embryos from natural matings are subject to numerous nuances, they nevertheless suggest that conclusions from mouse might not be able to be extended to all mammalian species.
It is not clear to us whether Reviewer 3’s comment implies that we have, at some point in the manuscript, generalized conclusions obtained in mice to other mammalian species—which we have not—or whether it is simply a general, common-sense remark with which we fully agree: that findings established in one species cannot, by default, be assumed to apply to another.
We would like to emphasize that throughout the manuscript, we have taken care to restrict our interpretations and conclusions to the mouse model, and we have avoided any unwarranted extrapolation to other species.
To definitively close this matter—if there is indeed a matter—we have added the following clarifying statements in the revised version of the manuscript:
In the introduction, second paragraph (pp. 2–3):"The variability across mammalian species in both the rate of fertilized oocytes with additional spermatozoa in their PVS (from 0 to more than 80%) after natural mating and the number of spermatozoa present in the PVS of these oocytes (from 0 to more than a hundred) suggests that the time for completion of the penetration block and thus its efficiency to prevent polyspermy can vary significantly between species."
At the end of the preamble to the Results section (p. 4):"This experimental study was conducted in mice, which are the most widely used model for studying fertilization and polyspermy blocks in mammals. While there are many interspecies similarities, the findings presented here should not be directly extrapolated to humans or other mammalian species without species-specific validation."
In the Conclusion, the first sentence is (p.22) : “This study sheds new light on the complex mechanisms that enable fertilization and ensure monospermy in mouse model.”
Within the Conclusion section, among the perspectives of this work (p. 22):"In parallel, comparative studies in other mammalian species will be needed to assess the generality of the PVS-block and its contribution relative to the membrane-block and ZP-blocks, as well as the generality of the mechanical role played by flagellar beating and ZP mechanical constraint in membrane fusion."
(4) Results, page 4 - It is very valuable that the authors clearly define what they mean by a penetrating spermatozoon and a fertilizing spermatozoon. However, they sometimes appear not to adhere to these definitions in other parts of the manuscript. An example of this is on page 10; the description of penetration of spermatozoon seems to be referring to membrane fusion with the oocyte plasma membrane, which the authors have alternatively called "fertilizing" or fertilization - although this is not entirely clear. The authors should go through all parts of the manuscript very carefully and ensure consistent use of their intended terminology.
Overall, while these definitions on page 4 are valuable, it is still recommended that the authors explicitly state when they are addressing penetration of the ZP and fertilization via fusion of the sperm with the oocyte plasma membrane. This help significantly in comprehension by readers. An example is the section header in the middle of page 9 - this could be "Spermatozoa can penetrate the ZP after the fertilization, but have very low chances to fertilize."
We chose to define our use of the term penetration at the beginning of the Results section because, as readers of fertilization studies, we have encountered on multiple occasions ambiguity as to whether this term was referring to sperm entry into the perivitelline space following zona pellucida traversal, or to the fusion of the sperm with the oolemma. To avoid such ambiguity, we were particularly careful throughout the writing of our original manuscript to use the term penetration exclusively to describe sperm entry into the PVS. The terms fertilizing and fusion were reserved specifically for membrane fusion between the gametes. However, as occasional lapses are always possible, we followed Reviewer 3’s recommendation and carefully re-examined the entire manuscript to ensure consistent use of our intended terminology. We did not identify any inconsistencies, including on page 10, which was cited as an example by Reviewer 3. We therefore confirm that, in accordance with our predefined terminology, all uses of the term penetration, on that page and anywhere else in our original manuscript, refer exclusively to sperm entry into the PVS and do not pertain to fusion with the oolemma.
That said, it is important that all readers— including those who may only consult selected parts of the article—are able to understand it clearly. Therefore, despite the potential risk of slightly overloading the text, Reviewer 3’s suggestion to systematically associate the term penetration with ZP seems to us a sound one. However, we have opted instead to associate penetration with PVS, as our study focuses on the timing of sperm penetration into the perivitelline space, rather than on the traversal of the zona pellucida itself. Accordingly, except in a few rare instances where ambiguity seemed impossible, we have systematically used the phrasing “penetration into the PVS” throughout the revised version of the manuscript.
Another variation of this is in the middle of page 9, where the authors use the terms "fertilization block" and "penetration block." These are not conventional terms, and venture into being jargon, which could leave some readers confused. The authors could clearly define what they mean, particularly with respect to "penetration block,"
This point has already been addressed in our response to Comment 1 from Reviewer 3. We invite Reviewer 3 to refer to that response.
This extends to other portions of the manuscript as well, such as Figure 2C, with the label on the y-axis being "Time after fertilization." It seems that what the authors actually observed here was the cessation of sperm tail motility. (It is not evident they they did an assessment of sperm-oocyte fusion here.)
Regarding Figure 2C (original version), it has been merged with Figure 2B (original version) to form a single figure (Figure S2D), now included in Supplementary Information SI2. This new figure retains all the information originally presented in Figure 2C and indicates the time axis origin as the time when oscillatory movements of the sperm cease.
That said, for the reasons detailed in our response to Reviewer 1 and in the Materials and Methods, we explain why it is legitimate to use the cessation of sperm head oscillations on the oolemma as a marker for the timing of the fusion event. We invite the reviewers to refer to that response for a full explanation of our rationale.
(5) Several points that the authors try to make with several pieces of data do not come across clearly in the text, including Figure 2 on page 6, Figure 4 on page 9, and the various states utilized for the statistical treatment, "post-first penetration, post-first fertilization, no fertilization, penetration block and polyspermy block" on page 10. Either re-writing and clearer definitions'explanations are needed, and/or schematic illustrations could be considered to augment re-written text. Illustrations could be a valuable way present the intended concepts to readers more clearly and accurately. For example, Figure 4 and the associated text on page 9 get particularly confusing - although this sounds like a quite impressive dataset with observations of 138 sperm. Illustrations could be helpful, in the spirit of "a picture is worth 1000 words," to show what seem to be three different situations of sequences of events with the sperm they observed. Finally, the text in the Results about the 138 sperm is quite difficult to follow. It also might help comprehension to augment the percentages with the actual numbers of sperm - e.g., is 48.6% referring 67 of the total 138 sperm analyzed? Does the 85.1% refer to 57 of these 67 sperm?
Figure 2 in the original version of our manuscript concerns sperm engulfment and PB2 extrusion. As already mentioned in our response to Reviewer 1, the characterization of sperm engulfment and PB2 extrusion kinetics is highly relevant to the analysis of the penetration and fusion blocks. However, we agree that its presence in the main text may distract the reader from the main focus of the study. Therefore, this figure and the associated text have been moved to the Supplementary Information in the revised manuscript (SI 2, pages 26–27).
Regarding Figure 4 (original version), in response to Reviewer 3’s concern about the difficulty in grasping the message conveyed in its three graphs and associated text we have completely rethought the way these data are presented. Since the three graphs of Figure 4 were directly derived from the experimental timing data of sperm entry in the PVS and fusion with the oolemma in fertilized oocytes (originally shown in Figure 3A), we have combined them into a single figure in the revised manuscript: Figure 3 (page 8). This new Figure 3 now comprises three components:
- Figure 3A remains unchanged from the original version and shows the timing of sperm penetration and fusion in fertilized oocytes. Each sperm category (fused or non-fused , penetrated in the PVS before fusion or after fusion) is represented using a color code clearly explained in the main text (last paragraph of page 7).
- Figure 3B focuses specifically on the first spermatozoon to penetrate the PVS of each oocyte. It reports how many of these first-penetrating spermatozoa succeeded in fusing versus how many failed to do so, highlighting that being the first to arrive is not sufficient for fusion—other factors are involved. This is explained simply in the first paragraph of page 9.
- Figure 3C considers all spermatozoa that entered the PVS of fertilized oocytes, classifying them into three categories: those that penetrated the PVS before fertilization, those that did so after fertilization, and those for which the timing could not be precisely determined. Such classification makes it apparent that the number of spermatozoa penetrating before and after fertilization is of the same order of magnitude, indicating that fertilization is not very effective at preventing further sperm entry into the PVS for the duration of our observations (~4 hours). To facilitate the identification of these three categories, the same color code used in Figure 3A is applied. In addition, within each category, the number of spermatozoa that successfully fused are indicated in black. This allows the reader to quickly assess the fertilization probability for each category—high for sperm entering before fertilization, very low or null for those entering after fertilization. This analysis shows that fertilization is far more effective at blocking sperm fusion than at blocking sperm penetration. This is clearly explained in the second paragraph of page 9. Regarding__ statistical analysis__, as already mentioned in our responses to Reviewers 1 and 2, this section has been rewritten to improve clarity and readability. The notation has also been significantly simplified. To improve the overall fluidity of the text related to the statistical analysis, Figure 3B (original version), which presented the timing of penetration into the perivitelline space of oocytes that remained unfertilized, along with its associated statistical analysis previously in Figure 5B), have been revised and transferred together in a single Figure S1 of the Supplementary Information (SI1, pages 26; now Figures S1A and S1B).
(6) Introduction, page 2 - it is inaccurate to state that only diploid zygotes can develop into a "new being." Triploid zygotes typically fail early in develop, but can survive and, for example, contribute to molar pregnancies. Additionally, it would be beneficial to be more scientifically precise term than saying "development into a new being." This is recommended not only for scientific accuracy, but also due to current debates, including in lay public circles, about what defines "life" or human life.
In response to Reviewer 3’s comment, we no longer state in the revised version of the manuscript that only diploid zygotes can develop into a new being. We have modified our wording as follows, on page 2, second paragraph: “In mammals, oocytes fertilized by more than one spermatozoon cannot develop into viable offspring.”
(7) Introduction, page 2 - The mammalian sperm must pass through three layers, not just two as stated in the first paragraph of the Introduction. The authors should include the cumulus layer in this list of events of fertilization.
The sentence from the introduction from the original manuscript mentioned by Reviewer 3 was: “To fertilize, a spermatozoon must successively pass two oocyte’s barriers.” This statement is accurate in the sense that the cumulus cell layer is not part of the oocyte itself, unlike the two oocyte’s barriers: the zona pellucida and the oolemma. Moreover, the traversal of the cumulus layer is not within the scope of our study, unlike the traversal of the zona pellucida and fusion with the oolemma. However, it is also correct that in our study the spermatozoa have passed through the cumulus layer before reaching the oocyte. Therefore, in response to Reviewer 3’s comment, we have revised the sentence to clarify this point as follows:
“Once a spermatozoon has passed through the cumulus cell layer surrounding the oocyte, it still must overcome two oocyte’s barriers to complete fertilization.”
(8) Introduction, page 2 - While there is evidence that zinc is released from mouse egg upon fertilization, the evidence is not convincing or conclusive that zinc is released from cortical granules or via cortical granule exocytosis.
To better highlight the rationale, storyline, and scope of our study, the introduction has been thoroughly streamlined. In this context, the section discussing the cortical reaction and zinc release seemed more appropriate in the Discussion, specifically within the paragraph titled “Relationship between the penetration block and the ZP-block.”
To address the uncertainty raised by Reviewer 3 regarding the origin of the zinc spark release, we have rephrased this part as follows:
“The fertilization-triggered processes responsible for the changes in ZP properties are generally attributed to the cortical reaction—a calcium-induced exocytosis of secretory granules (cortical granules) present in the cortex of unfertilized mammalian oocytes—and to zinc sparks. As a result, proteases, glycosidases, lectins, and zinc are released into the perivitelline space (PVS), where they act on the components of the zona pellucida. This leads to a series of modifications collectively referred to as ZP hardening or the ZP-block”.
(9) The authors inaccurately state, "only if monospermic multi-penetrated oocytes are able to develop normally, which to our knowledge has never been proven in mice" (page 4) - This was demonstrated with the Astl knockout, assuming that the authors use of "multi-penetrated oocytes" here refers to the definition of penetration that they use, namely penetrating the ZP. This also is one of the instances where the authors contradict themselves, as they note the results with this knockout on page 18.
Thank you for bringing this point to our attention. Nozawa et al. (2018) found that female mice lacking ovastacin (Astl)—the protease released during the cortical reaction that plays a key role in rendering the zona pellucida impenetrable—are normally fertile. They also reported that oocytes recovered from these females after mating were monospermic, despite the consistent presence of additional spermatozoa in the perivitelline space. We can indeed consider that taken together these findings demonstrate that the presence of multiple spermatozoa in the PVS does not impair normal development, as long as the oocyte remains monospermic. In our study, we re-demonstrated this in a different way (by reimplantation of monospermic oocytes with additional spermatozoa in their PVS) in a more physiological context of WT oocytes, but we agree that we cannot state: “which to our knowledge has never been proven in mice.” This part of the sentence has therefore been removed. In the revised version of the manuscript, the sentence is now formulated in the first paragraph of page 5 as follows: “However, the contribution of the fusion block to prevent polyspermy has physiological significance only if monospermic oocytes with additional spermatozoa in their PVS can develop into viable pups.”
Minor comments:
There are numerous places where this reader marked places of confusion in the text. A sample of some of these:
We will indicate hereinafter how we have modified the text in the specific examples provided by Reviewer 3. Beyond these, however, we would like to emphasize that we have thoroughly revised the entire manuscript to improve clarity and precision.
Page 4 - "continuously relayed by other if they detach" - don't know what this means
Replaced now p 5 by “can be replaced by others if they detach”
Page 6 - "hernia" - do the authors mean "protrusion" on the oocyte surface?
The paragraph from the Results section in question has now been moved to the Supplementary Information, on pages 26 and 27. The term hernia has been systematically replaced with protrusion, including in the Materials and Methods section on page 24.
Page 10 - "penetration of spermatozoa in the PVS falls down" - don't know what this means
Falls down has been removed from the new version and replaced with decreases
Page 12 - "spermatozoa linked to the oocyte ZP" - not clear what "linked" means here
Replaced now page 16 by “spermatozoa bound to the oocyte ZP”
Page 14 - "by dint of oscillations" - don't know what this means
Replaced now page 10 by “the persistent flagellum movements”
Specifics for Materials and Methods:
Exact timing of females receiving hCG and then being put with males for mating - assume this was immediate but this is an important detail regarding the timing for the creation of embryos in vivo.
That is correct: females were placed with males for mating immediately after receiving hCG. This clarification has been added in the revised version of the manuscript.
Please provide the volumes in which inseminations occurred, and how many eggs were placed in this volume with the 10^6 sperm/ml.
The number of eggs may vary from one cumulus–oocyte complex to another. It is therefore not possible to specify exactly how many eggs were inseminated. However, we now indicate on page 23 the number of cumulus–oocyte complexes inseminated (4 per experiment), the volume in which insemination was performed (200 mL), and the sperm concentration used 106 sperm/mL.
**Referees cross-commenting**
I concur with Reviewer 1's comment, that the 'challenging prior dogma' about the first sperm not always being the one to fertilize the egg is too strong. As Reviewer 1 notes, "it had been observed before that it is not necessarily the first sperm that gets through the ZP that fertilizes the egg." I even thought about adding this comment to my review, although held off (I was hoping to find references, but that was taking too long).
Please refer to our response to Reviewer 1 regarding this point.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Summary:
This study by Dubois et al. utilizes live-cell imaging studies of mouse oocytes undergoing fertilization. A strength of this study is their use of three different conditions for analyses of events of fertilization: (1) eggs undergoing fertilization retrieved from females at 15 hr after mating (n = 211 oocytes); (2) cumulus-oocyte complexes inseminated in vitro (n = 220 oocytes), and (3) zona pellucida (ZP)-intact eggs inseminated in vitro, transferred from insemination culture once sperm were observed bound to the ZP for subsequent live-cell imaging (93 oocytes). This dataset and these analyses are valuable for the field of fertilization biology. Limitations of this manuscript are challenges arise with some conclusions, and the presentation of the manuscript. There are some factual errors, and also some places where clearer explanations should to be provided, in the text and potentially augmented with illustrations to provide more clarity on the models that the authors interpret from their data.
Major comments:
The authors are congratulated on their impressive collection of data from live-cell imaging. However, the writing in several sections is challenging to understand or seems to be of questionable accuracy. The lack of accuracy is suspected to be more an effect of overly ambitious attempts with writing style, rather than to mislead readers. Nevertheless, these aspects of the writing should be corrected. There also are multiple places where the manuscript contradicts itself. These contradictions should be corrected. Finally, there are factual points from previous studies that need correction.
Second, certain claims and the conclusions as presented are not always clearly supported by the data. This may be connected to the issues with writing style, word and phrasing choices, etc. The conclusions could be expressed more clearly, and thus may not require additional experiments or analyses to support them. The authors might also consider illustrations as ways to highlight the points they wish to make. (Figure 7 is a strong example of how they use illustrations to complement the text).
Specific comments:
This also is an instance of where the authors contradict themselves in the manuscript, stating, "the membrane block and the ZP block are established in approximatively the same time frame" (third paragraph of Introduction). This statement is indeed accurate, unlike the reference to a fast block to polyspermy in mammals.<br /> 2. The authors aim to make the case that events occurring in the perivitelline space (PVS) prevent polyspermic fertilization, but the data that they present is not strong enough to make this conclusion. Additional experiments would optional for this study, but data from such additional experiments are needed to support the authors' claims regarding these functions in fertilization. Without additional data, the authors need to be much more conservative in interpretations of their data. The authors have indeed observed phenomena (the presence of CD9 and JUNO in the PVS) that could be consistent with a molecular basis of a means to prevent fertilization by a second sperm. However, the authors would need additional data from additional experimental studies, such as interfering with the release of CD9 and JUNO and showing that this experimental manipulation leads to increased polyspermy, or creating an experimental situation that mimics the presence of CD9 and JUNO (in essence, what the authors call "sperm inhibiting medium" on page 20) and showing that this prevents fertilization.
A major section of the Results section here (starting with "The consequence is that ... ") is speculation. Rather than be in the Results section, this should be in the Discussion. The language should be also softened regarding the roles of these proteins in the perivitelline space in other portions of the manuscript, such as the abstract and the introduction.
Finally, the authors should do more to discuss their results with the results of Miyado et al. (2008), which interestingly, posited that CD9 is released from the oocytes and that this facilitates fertilization by rendering sperm more fusion-competent. There admittedly are two reports that present data that suggest lack of detection of CD9-containing exosomes from eggs (as proposed by Miyado et al.), but nevertheless, the authors should put their results in context with previous findings. 3. Many of the authors' conclusions focus on their prior analyses of sperm interaction - beautifully illustrated in Figure 7. However, the authors need to be cautious in their interpretations of these data and generalizing them to mammalian fertilization as a whole, because mouse and other rodent sperm have sperm head morphology that is quite different from most other mammalian species.
In a similar vein, the authors should be cautious in their interpretations regarding the extension of these results to mammalian species other than mouse, given data on numbers of perivitelline sperm (ranging from 100s in some species to virtually none in other species), suggesting that different species rely on different egg-based blocks to polyspermy to varying extents. While these observations of embryos from natural matings are subject to numerous nuances, they nevertheless suggest that conclusions from mouse might not be able to be extended to all mammalian species.<br /> 4. Results, page 4 - It is very valuable that the authors clearly define what they mean by a penetrating spermatozoon and a fertilizing spermatozoon. However, they sometimes appear not to adhere to these definitions in other parts of the manuscript. An example of this is on page 10; the description of penetration of spermatozoon seems to be referring to membrane fusion with the oocyte plasma membrane, which the authors have alternatively called "fertilizing" or fertilization - although this is not entirely clear. The authors should go through all parts of the manuscript very carefully and ensure consistent use of their intended terminology.
Overall, while these definitions on page 4 are valuable, it is still recommended that the authors explicitly state when they are addressing penetration of the ZP and fertilization via fusion of the sperm with the oocyte plasma membrane. This help significantly in comprehension by readers. An example is the section header in the middle of page 9 - this could be "Spermatozoa can penetrate the ZP after the fertilization, but have very low chances to fertilize."
Another variation of this is in the middle of page 9, where the authors use the terms "fertilization block" and "penetration block." These are not conventional terms, and venture into being jargon, which could leave some readers confused. The authors could clearly define what they mean, particularly with respect to "penetration block,"
This extends to other portions of the manuscript as well, such as Figure 2C, with the label on the y-axis being "Time after fertilization." It seems that what the authors actually observed here was the cessation of sperm tail motility. (It is not evident they they did an assessment of sperm-oocyte fusion here.) 5. Several points that the authors try to make with several pieces of data do not come across clearly in the text, including Figure 2 on page 6, Figure 4 on page 9, and the various states utilized for the statistical treatment, "post-first penetration, post-first fertilization, no fertilization, penetration block and polyspermy block" on page 10 . Either re-writing and clearer definitions'explanations are needed, and/or schematic illustrations could be considered to augment re-written text. Illustrations could be a valuable way present the intended concepts to readers more clearly and accurately. For example, Figure 4 and the associated text on page 9 get particularly confusing - although this sounds like a quite impressive dataset with observations of 138 sperm. Illustrations could be helpful, in the spirit of "a picture is worth 1000 words," to show what seem to be three different situations of sequences of events with the sperm they observed. Finally, the text in the Results about the 138 sperm is quite difficult to follow. It also might help comprehension to augment the percentages with the actual numbers of sperm - e.g., is 48.6% referring 67 of the total 138 sperm analyzed? Does the 85.1% refer to 57 of these 67 sperm?<br /> 6. Introduction, page 2 - it is inaccurate to state that only diploid zygotes can develop into a "new being." Triploid zygotes typically fail early in develop, but can survive and, for example, contribute to molar pregnancies. Additionally, it would be beneficial to be more scientifically precise term than saying "development into a new being." This is recommended not only for scientific accuracy, but also due to current debates, including in lay public circles, about what defines "life" or human life. <br /> 7. Introduction, page 2 - The mammalian sperm must pass through three layers, not just two as stated in the first paragraph of the Introduction. The authors should include the cumulus layer in this list of events of fertilization. 8. Introduction, page 2 - While there is evidence that zinc is released from mouse egg upon fertilization, the evidence is not convincing or conclusive that zinc is released from cortical granules or via cortical granule exocytosis.<br /> 9. The authors inaccurately state, "only if monospermic multi-penetrated oocytes are able to develop normally, which to our knowledge has never been proven in mice" (page 4) - This was demonstrated with the Astl knockout, assuming that the authors use of "multi-penetrated oocytes" here refers to the definition of penetration that they use, namely penetrating the ZP. This also is one of the instances where the authors contradict themselves, as they note the results with this knockout on page 18.
Minor comments:
There are numerous places where this reader marked places of confusion in the text. A sample of some of these:
Page 4 - "continuously relayed by other if they detach" - don't know what this means
Page 6 - "hernia" - do the authors mean "protrusion" on the oocyte surface?
Page 10 - "penetration of spermatozoa in the PVS falls down" - don't know what this means
Page 12 - "spermatozoa linked to the oocyte ZP" - not clear what "linked" means here
Page 14 - "by dint of oscillations" - don't know what this means
Specifics for Materials and Methods:
Exact timing of females receiving hCG and then being put with males for mating - assume this was immediate but this is an important detail regarding the timing for the creation of embryos in vivo.
Please provide the volumes in which inseminations occurred, and how many eggs were placed in this volume with the 10^6 sperm/ml.
Referees cross-commenting
I concur with Reviewer 1's comment, that the 'challenging prior dogma' about the first sperm not always being the one to fertilize the egg is too strong. As Reviewer 1 notes, "it had been observed before that it is not necessarily the first sperm that gets through the ZP that fertilizes the egg." I even thought about adding this comment to my review, although held off (I was hoping to find references, but that was taking too long).
This manuscript brings interesting new observations for the field of gamete and fertilization biology. For very obvious reasons, the understanding of mammalian fertilization has lagged behind the understanding of fertilization of species with external fertilization. Decades ago, developmental biologists first focused on studies of fertilization on gametes from species that release sperm and egg into water, either spontaneously or with relatively easy stimulation, and gametes that could be easily cultured and enabled to create embryos as researchers watched. Studies of mammalian fertilization have since caught up, with the elucidation of conditions that support in vitro fertilization in various mammalian species, most notably mouse as an experimental model.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Overall, this is a very interesting and relevant work for the field of fertilization. In general, the experimental strategies are adequate and well carried out. I have some questions and suggestions that should be considered before the work is published.
Understanding mammalian gamete fusion and polyspermy inhibition has not been fully achieved. The authors examined real time brightfield and confocal images of inseminated ZP-intact mouse oocytes and used statistical analyses to accurately determine the dynamics of the events that lead to fusion and involve polyspermy prevention under conditions as physiological as possible. Their kinetic observations in mice gamete interactions challenge present paradigms, as they document that the first sperm is not necessarily the one that fertilizes, suggesting the existence of other post-penetration fertilization factors. The authors find that the zona pellucida (ZP) block triggered by the cortical reaction is too slow to prevent polyspermy in this species. In contrast, their findings indicate that ZP directly contributes to the polyspermy block operating as a naturally effective entry barrier inhibiting the exit from the perivitelline space (PVS) of components released from the oocyte plasma membrane (OPM), neutralizing unwanted sperm fusion, aside from any block caused by fertilization. Furthermore, the authors unveil a new important ZP role regulating flagellar beat in fertilization by promoting sperm fusion in the PVS.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The manuscript "Key roles of the zona pellucida and perivitelline space in promoting gamete fusion and fast block to polyspermy inferred from the choreography of spermatozoa in mice oocytes" by Dr. Gourier and colleagues explores the poorly understood process of gamete fusion and the subsequent block to polyspermy by live-cell imaging of mouse oocytes with intact zona pellucida in vitro. The new component in this study is the presence of the ZP, which in prior studies of live-cell imaging had been removed before. This allowed the authos to examine contributions of the ZP to the block in polyspermy in relation to the timing of sperm penetrating the ZP and sperm fusing with the oocyte. By carefully analysing the timing of the cascade of events, the authors find that the first sperm that reaches the membrane of the mouse oocyte is not necessarily the one that fertilizes the oocytes, revealing that other mechanisms post-ZP-penetration influence the success of individual sperm. While the rate of ZP penetration remains constant in unfertilized oocytes, it decreases upon fertilization for subsequent sperm, providing direct evidence for the known 'slow block to polyspermy' provided by changes to the ZP adhesion/ability to be penetrated. Careful statistical analyses allow the authors to revisit the role of the ZP in preventing polyspermy: They show that the ZP block resulting from the cortical reaction is too slow (in the range of an hour) to contribute to the immediate prevention of polyspermy in mice. The presented analyses reveal that the ZP does contribute to the block to polyspermy in two other ways, namely by effectively limiting the number of sperm that reach the oocyte surface in a fertilization-independent manner, and by retaining components like JUNO and CD9, that are shed from the oocyte plasma membrane after fertilization, in the perivitelline space, which may help neutralize surplus spermatozoa that are already present in the PVS. Lastly, the authors report that the ZP may also contribute to channeling the flagellar oscillations of spermatozoa in the PVS to promote their fusion competence.
Major comments:
The authors provide a careful analysis of the dynamics of events, though the analyses are correlative, and can only be suggestive of causation. While this is a limitation of the study, it provides important analysis for future research. Moreover, by analysing also control oocytes without fertilization and the timing of events, the authors have in some instances clear 'negative controls' for comparison.
Some claims would benefit from rewording or rephrasing to put the findings better in the context of what is already known and what is novel: - the phrasing 'challenging prior dogma' might be too strong since it had been observed before that it is not necessarily the first sperm that gets through the ZP that fertilizes the egg (though I am afraid that I do not have any citations or references for this). However, given that in the field people generally think it is not necessarily and always the first sperm, the authors may want to consider weakening this claim. - I do think the cortical granule release could still contribute to the block to polyspermy though - as the authors here nicely show - at a later time-point only, and thus not the major and not the immediate block as previously thought. The wording in the abstract should therefore be adjusted (since it could still contribute...) - the finding that the ZP presents a natural effective barrier for sperm entry is not that novel (as suggested here) - there are mutants that prevent sperm from getting through the ZP and thus to the oocyte and those lead to sterility - release of OPM components - in the abstract it's unclear what the authors mean by this - in the results part it becomes clear. Please already make it clear in the abstract that it is the fertility factors JUNO/CD9 that could bind to sperm heads upon their release and thus 'neutralize' them? I would also recommend not referring to it as 'outer' plasma membrane (there is no 'inner plasma membrane'). Moreover, in the abstract please clarify that this release is happening only after fusion of the first sperm and not all the time. In the abstract it sounds as if this was a completely new idea, but there is good prior evidence that this is in fact happening (as also then cited in the results part) - maybe frame it more as the retention inside the PVS as new finding.
It is unclear to me what the relevance of dividing the post-fusion/post-engulfment into different phases as done in Fig 2 (phase 1, and phase 2) - also for the conclusions of this paper this seems rather irrelevant and overly complicated, since the authors never get back to it and don't need it (it's not related to the polyspermy block analyses). I would remove it from the main figures and not divide into those phases since it is distracting from the main focus.
For the statistical analysis, I am not sure whether the assumption "assumption that the probability distribution of penetration or fertilization is uniform within a given time window" is in fact true since the probability of fertilizing decreases after the first fertilization event.... Maybe I misunderstood this, but this needs to be explained (or clarified) better, or the limitation of this assumption needs to be highlighted. - Suggestion for additional experiments:
If I understood correctly, the onset of fusion in Fig 2C is defined by stopping of sperm beating? If it is by the sudden stop of the beating flagellum, this should be confirmed in this situation (with the ZP intact) that it correctly defines the time-point of fusion since this has not been measured in this set-up before as far as I understand. In order to measure this accurately, the authors will need to measure this accurate to be able to acquire those numbers (of time from fusion to end of engulfment), e.g. by pre-loading the oocyte with Hoechst to transfer Hoechst to the fusing sperm upon membrane fusion.
Fig 8: 2 comments - To better show JUNO/CD9 pre-fusion attachment to the oocyte surface and post-fusion loss from the oocyte surface (but persistence in the PVS), an image after removal of the ZP (both for pre-fertilization and post-fertilization) would be helpful - the combination of those images with the ones you have (ZP intact) would make your point more visible. - You show that the heads of spermatozoa post fusion are covered in CD9 and JUNO, yet I was missing an image of sperm in the PVS pre-fertilization (which should then not yet be covered).
Minor comments:
Overall, this manuscript provides very interesting and carefully obtained data which provides important new insights particularly for reproductive biology. I applaud the authors on first establishing the in vivo conditions (how often do multiple sperm even penetrate the ZP in vivo) since studies have usually just started with in vitro condition where sperm at much higher concentration is added to isolated oocyte complexes. Thank you for providing an in vivo benchmark for the frequency of multiple sperm being in the PVS. While this frequency is rather low (somewhat expectedly, with 16% showing 2-3 sperm in the PVS), this condition clearly exists, providing a clear rationale for the investigation of mechanisms that can prevent additional sperm from entering.
My own expertise is experimentally - thus I don't have sufficient expertise to evaluate the statistical methods employed here.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer #1 (Evidence, reproducibility and clarity (Required)):
The authors use Dyngo-4a, a known Dynami inhibitor to test its influence on caveolar assembly and surface mobility. They investigate, whether it incorporates into membranes with Quartz-Crystal Microbalance, they investigate how it is organized in membranes using simulations. Finally, they use lipid-packing sensitive dyes to investigate lipid packing in the presence of Dyngo-4a, membrane stiffness using AFM and membrane undulation using fluorescence microscopy. They also use a measure they call "caveola duration time" to claim that something happens to caveolae after Dyngo-4a addition and using this parameter, they do indeed see an increase in it in response to Dyngo-4a, which is reduced back to the baseline after addition of cholesterol.
Overall, the authors claim: 1) Dyngo-4a inserts into the membrane and this 2) results in "a dramatic dynamin-independent inhibition of caveola scission". 3) Dyngo-4a was inserted and positioned at the level of cholesterol in the bilayer and 4) Dyngo-4a-treatment resulted in decreased lipid packing in the outer leaflet of the plasma membrane 5) but Dyngo-4a did not affect caveola morphology, caveolae- associated proteins, or the overall membrane stiffness 6) acute addition of cholesterol counteracts the block in caveola scission caused by Dyngo-4a
Overall, in this reviewers opinion, claims 1, 3, 4, 5 are well-supported by the presented data from electron and live cell microscopy, QCM-D and AFM. However, there is no convincing assay for caveolar endocytosis presented besides the "caveola duration" which although unclearly described seems to be the time it takes in imaging until a caveolae is not picked up by the tracking software anymore in TIRF microscopy. Since the main claim of the paper is a mechanism of caveolar endocytosis being blocked by Dyngo-4a, a true caveolar internalization assays is required to make this claim. This means either the intracellular detection of not surface connected caveolar cargo or the quantification of caveolar movement from TIRF into epifluorescence detection in the fluorescence microscope. Otherwise, the authors could remove the claim and just claim that caveolar mobility is influenced.
Response: We thank the reviewer for the nice constructive comments, and we very much appreciate the positive critique. We have now included a FRAP experiment of endocytic Cav1-GFP supporting the effect on internalization. In addition, we are currently preforming CTxB HRP experiments to quantify the number of caveolae at PM using EM but due to reasons out of our control we have not managed to finish these on time, they will be included in the manuscript once they are ready in hopefully not too long.
Reviewer #1 (Significance (Required)):
A number of small molecule inhibitors for the GTPase dynamics exist, that are commonly used tools in the investigation of endocytosis. This goes as far that the use of some of these inhibitors alone is considered in some publications as sufficient to declare a process to be dynamin-dependent. However, this is not correct, as there are considerable off-target effects, including the inhibition of caveolar internalization by a dynamin-independent mechanism. This is important, as for example the influence of dynamin small molecule inhibitors on chemotherapy resistance is currently investigated (see for example Tremblay et al., Nature Communications, 2020).
The investigation of the true effect of small molecules discovered as and used as specific inhibitors and their offside effects is extremely important and this reviewer applauds the effort. It is important that inhibitors are not used alone, but other means of targeting a mechanism are exploited as well in functional studies. The audience here thus is besides membrane biophysicists interested in the immediate effect of the small molecule Dyngo-4a also cell biologists and everyone using dynamic inhibitors to investigate cellular function.
__Reviewer #2 (Evidence, reproducibility and clarity (Required)): __
This manuscript uses the small molecule dynamin inhibitors dynasore and dyngo to show that in dynamin triple knockout cells that these inhibitors impact lipid packing and organization in the plasma membrane. Data showing that dyngo affects caveolin dynamics using tirf microscopy is also shown and is interpreted to reflect inhibition of caveolae scission from the membrane.
This data showing that dyngo and dynasore target membrane order is quite compelling and argues that the effects of these inhibitors is not dynamin specific and that inhibition of endocytosis by these small molecule inhibitors is dynamin-independent. The in vitro and in vivo data they provide is convincing.
Similarly, the data showing that dynasore and dyngo affect caveolin dynamics and clathrin endocytosis (transferrin) is quite convincing and argues that altered lipid packing is impacting membrane dynamics at the plasma membrane. What is less convincing is the conclusion is that dyngo is preventing caveolae scission from the membrane. Study of caveolae endocytosis is based on a TIRF assay that has inherent limitations: - Caveolae are defined as bright cav1-positive spots in diffraction limited TIRF and their disappearance presumed to be endocytic events. Cav1 spots are presumed to be caveolae but the authors do not consider that they may be flat non-caveolar oligomers. The diffraction limited TIRF approach interprets the large structures as caveolae but evidence to that effect is lacking.
Response: This is a valid comment and to address this we have now included data showing colocalization of cavin1 and EHD2 to the Cav1-GFP spots. We can however not determine if they are flat or invaginated. We do have extensive experience imaging caveolae using TIRF microscopy and carefully chose cells that display low expression of fluorescently labelled caveolin to avoid non-caveolar structures.
- The analysis (and the diagram presented in figure 4) considers that caveolae can either diffuse laterally in the membrane or internalize and does not consider that caveolae can flatten and possibly fragment in the membrane. Is it not possible that loss of Cav1 spots is a fragmentation event and not necessarily a scission event?
Response: This is a good question, yet, fragmentation and disassembly would result in shorter track durations and this is not what is observed in data. We have now also included data showing that cavin1 is persistently associated with the Cav1 spots identified as caveolae during Dyngo-4a treatment indicating that these are caveolae. Furthermore, IF stainings showing colocalization of Cav1GFP with cavin1 or EHD2 after Dyngo-4a treatment have also been added. We have now also expanded on the different interpretations of the data in the results section.
- The analysis is based on overexpression of Cav1-GFP that may alter the stoichiometry between Cav1 and cavin1 such that while caveolae may be expressed, larger non-caveolar structures may accumulate.
Response: Yes, this is correct, we have specifically imaged cell expressing low levels of Cav1-GFP to avoid accumulated non-caveolar structures that can be spotted in cells with high expression.
- Cav1 has been shown to be internalized via the CLIC pathway (Chaudary et al, 2014) and if dyngo is impacting clathrin then maybe it is also impacting CLIC endocytosis and thereby Cav1 endocytosis via this pathway?
Response: Dyngo-4a has been shown to not affect CLIC endocytosis (McCluskey et al., 2013) and in our data we do not see internalization following Dyngo-4a treatment.
- The longer Cav1 TIRF track time and shorter displacement with dyngo is consistent with inhibition of caveolae scission. However, as the authors discuss, could not reduced membrane undulations due to dyngo's impact on membrane order be responsible for the longer tracks? Alternatively, perhaps the altered lipid packing is corralling Cav1 movement and reducing non-caveolar Cav1 endocytosis, resulting in shorter tracks of longer duration? The proposed interaction of dyngo with cholesterol could prevent scission but also stabilize large (flat?) Cav1 oligomers in the membrane, perhaps reducing Cav1 oligomer fragmentation.
Response: We completely agree that membrane undulations contribute to instability of the TIRF-field and therefore disruption of cav1-GFP tracks as we discuss in the results section and have been described in previous work (Larsson et al., 2023). Yet, we have also shown that internalization of caveolae results in shorter tracks (Hubert et al., 2020; Larsson et al., 2023; Mohan et al., 2015). Furthermore, the tracked Cav1-GFP spots are persistently positive for cavin1 both with and without Dyngo-4a treatment showing that the majority do not disassemble become internalized by other pathways. Additionally, the added IF stainings after 30 min Dyngo-4a treatment also show that the Cav1-GFP spots remain positive for cavin1 and EHD2 just as ctrl-treated cells.
My point here is not to discredit the data but only to suggest that the TIRF approach used is an indirect measure of caveolae scission from the membrane that requires substantiation using other approaches.
Response: We appreciate these comments and have tried to address these by adding new data and discussions on the interpretation of the tracking data in the results section.
Dyngo is certainly generally affecting lipid packing via cholesterol and thereby affecting Cav1 dynamics in the plasma membrane. The claim of caveolae scission should be qualified and alternative possibilities considered and discussed. If the authors persist in arguing that dyngo is affecting caveolae scission then the effect should be substantiated by accumulation of caveolae by quantitative EM and high spatial and temporal resolution imaging of Cav1 and cavin1 to define the endocytic events. As the latter represents a new, and potentially very challenging, line of experimentation, I would suggest that it is beyond the scope of the current study. As indicated above the additional experiments are not necessary and qualification of the claims would be sufficient.
-Response: We have now included a FRAP experiment of endocytic Cav1-GFP supporting the effect on internalization. We are also currently preforming CTxB HRP experiments to quantify the number of caveolae at the PM using EM but due to reasons out of our control we have not managed to finish these on time, they will be included in the manuscript once they are ready in hopefully not too long.
Other points
Figure 1C - Cav1 positive spots cannot be interpreted to be caveolae from diffraction limited confocal images. Same comment applies to Fig 4G - caveola? duration.
-Response: We completely agree with this and that the claims should be qualified. We have added IF stainings showing that the Cav1-GFP structures are also positive for cavin1. We have now clarified that we cannot distinguish between flat or different curved states of caveolae using this methodology. We have also changed the labelling of Fig. 4G.
Figure 4C - it is not clear why this EM data is not quantified - for both the number of caveolae and clathrin coated pits - as this would help clarify the interpretation of the effect reported.
-Response: We are currently preforming CTxB HRP experiments to quantify the number of caveolae using EM but due to reasons out of our control we have not managed to finish these on time, they will be included in the manuscript once they are ready in hopefully not too long.
Figure 4D - the AFM experiments should perhaps be repeated as the non-significant effect of dyngo on the Young's modulus may be a result of insufficient n values. -Response: We would like to clarify that to ensure the robustness of our AFM measurements, we performed the experiments with sufficient biological and technical replicates. Specifically, each data point shown in Figure 4D represents a Young’s modulus value averaged from approximately sixty force-distance curves per cell. For each condition, we collected force-distance maps on eight to nine individual cells, obtained from two separate petri dishes per day. We repeated this process on two independent days. In total, we analysed thirty-one cells for the DMSO control and thirty-three cells for the Dyngo-4a treatment. We performed the “student’s t-test with Welch’s correction” to access the statistical significance between the two conditions, as described in the main text. We believe that the sample size and statistical approach are sufficient to support the conclusions presented. Furthermore, we also analysed cell stiffness by calculating the slope of the linear portion of the force-distance curves. This analysis also did not reveal any statistically significant differences between the conditions (data not shown), further supporting our conclusion that Dyngo-4a treatment does not significantly alter the Young’s modulus under our experimental setup (or conditions).
Reviewer #2 (Significance (Required)):
This data showing that dyngo and dynasore target membrane order is quite compelling and argues that the effects of these inhibitors is not dynamin specific and that inhibition of endocytosis by these small molecule inhibitors is dynamin-independent. The in vitro and in vivo data they provide is convincing.
Similarly, the data showing that dynasore and dyngo affect caveolin dynamics and clathrin endocytosis (transferrin) is quite convincing and argues that altered lipid packing is impacting membrane dynamics at the plasma membrane. What is less convincing is the conclusion is that dyngo is preventing caveolae scission from the membrane.
__Reviewer #3 (Evidence, reproducibility and clarity (Required)): __
Larsson et al present experimental and computational data on the role of Dyngo4a (a compound that was developed to inhibit dynamin) on the dynamics of caveolae. The manuscript mostly documents effects of Dyngo on caveolae, with one experiment to suggest a mechanism for this result. This one rather unconvincing result forms the focus of the manuscript contributing to a disconnect between the data and the presentation. Additionally, there are concerns with data interpretation. The writing could also benefit from revision to address grammar mistakes, strengthen referencing, and increase precision. Overall, the manuscript requires substantial revisions before being considered for publication. The central claim, in particular, needs stronger evidence to support the proposed mechanism. -Response: We thank the reviewer for the thorough review and for experimental suggestions that we believe has strengthened our data further.
Significant issues (in approximate order of importance): 1. The data supporting the central mechanistic explanation appears limited. There is no evidence that Dyngo remains in one leaflet
Response:The simulations show that the energy barrier for moving in between bilayers is very high. Furthermore, simulations of C-Laurdan has shown that it does not readily flip in between membrane leaflets (Barucha-Kraszewska et al., 2013) supporting that it reports on the outer lipid leaflet when added to cells. We have however now changed this and state that Dyngo-4a decreased the lipid order in the plasma membrane.
the GP of the PM is very low compared to previous measurements,
Response: The absolute GP-values will vary between setups depending on what filters are used so they are not comparable between laboratories. What is of importance is that we found a significant change in the relative GP-values in cells treated with Dyngo-4a and control cells. It is this change that we report. We have not performed any GP-measurements on this cell type earlier so it is unclear what previous measurements reviewer #3 are referring to.
effects on other membranes are not explored,
Response: The order of the intracellular membranes is as expected lower than that of the plasma membrane. Differentiating different intracellular membranes of interest like endocytotic vesicles from other intracellular membranes would be very difficult but, more importantly, our study is focused on what is happening in the plasma membrane where caveolae reside and would be of minor interest for plasma membrane dynamics.
dynamin-directed effects of Dyngo are not considered,
Response: In the discussion section we discuss the difficulties with disentangling dynamin-direct and indirect effects.
The QCM-D measurements and claims require explanation as several aspects remains unclear. In Fig S2, the 'softness' (what does this mean?) changes by 4-fold with DMSO alone (what does this mean?), then fractionally more with Dyngo. Then fractionally more again when Dyngo is removed (why?). Then it remains somewhat higher when both Dyngo and DMSO are removed, which is somehow interpreted as Dyngo remaining in the bilayer, but not DMSO.
Response:We understand the confusion of the reviewer and hope our explanations provide clarity. QCM-D measurements are based on an oscillating quartz crystal sensor. Specifically, alterations in oscillation frequency (ΔF) and the rate of energy dissipation from the sensor surface (ΔD) are what is measured. Allowing the measurement of: 1) materials adsorbing to the sensor surface, 2) changes in the viscoelastic properties of a solution in contact with the sensor surface, 3) changes in the material adsorbed to the sensor surface upone exposure to different solutions. The ratio of ΔD/-ΔF reports the mechanical softness or rigidity of an adsorbed material, in this case the SLB.A “buffer shift” is the term used when there is not an adsorption to the sensor surface, but rather an effect from altering the solution above the sensor surface. One reason is because different solutions can have different densities (e.g., a DMSO-buffer mixture vs buffer alone), which impacts the oscillations of the sensor. It was observed that the DMSO-buffer mixture alone gave a large buffer shift in comparison to the adsorption of the Dyngo-4a into the SLB, thereby muddling the data interpretation. Thus, in Fig. S2 the system was first equilibrated with the DMSO-buffer mixture prior to addition of the Dyngo-4a solution to allow for clearer visualization of the two events. In QCMD to assess if something has made a permeant change to the system you change back to the solutions used before the addition, thus first we washed with a DMSO-Buffer mixture followed by buffer alone. Control experiments were carried out in which no Dyngo-4a was added (also shown in Fig. S2). The control shows the same “buffer shift” from the DMSO-buffer mixture occurs in both systems and that upon returning to a buffer only condition there is no permanent change to the system caused from exposure to the DMSO. In contrast, once the system that received Dyngo-4a is changes back to a buffer only system we see that mass has been added to the system (ΔF) with little change to the dissipation (ΔD), thereby resulting in a lower ratio of ΔD/-ΔF, which is to say that the SLB after the adsorption of Dyngo-4a was more rigid that the SLB without Dyngo-4a.
These interpretations are difficult to grasp, as the authors seem to be implying simple amphiphilic partitioning into the membrane, which should all be removable by efficient washing.
Response: Amphiphilic partitioning is not fully reversible by “efficient washing” it depends on partitioning coefficients.
I do not doubt that this compound interacts with membranes, but the quantifications appear ambiguous. A bilayer with 16 mol% (or worse, 30% if all in one leaflet) Dyngo is very unlikely (to remain a bilayer). Even if such a bilayer was conceivable, the authors are claiming an ADDITION of Dyngo that would INCREASE the area of one leaflet by 30%, which needs explanation as it appears unlikely.
-Response: We understand that in our attempt provide numbers in the results section for the amount of binding observed in QCM-D, this can easily be interpreted as this is what is observed to insert into the PM. However, as discussed in the discussion, we also see aggregations of Dyngo-4a that associate with the membrane in the simulations which likely could contribute to the binding observed in QCM-D prior to washing. The precise amount of membrane inserted Dyngo-4a is difficult to measure as we discuss in the text. In order to make this clearer, we have now moved all these details to the discussion section where we elaborate on this. Furthermore, since Dyngo-4a, like cholesterol, is intercalating in between the head groups of the lipids the area would not increase in direct proportion to the mol%.
Also, there are no replicates shown, so unclear how reproducible these effects are?
Response: For clarity, only single experiments are shown. However, multiple experiments were performed and the range in measured values for 3 technical repeats can be observed in the standard deviations found in the main text (e.g., 6 ± 2 mol%).
The simulations are insufficiently described and difficult to interpret. How big are these systems? Why do the figures show the aqueous system with lateral boundaries?
Response: There are no explicit boundaries used in the simulations, periodic boundary conditions are applied in all three dimensions. The lateral boundaries observed in the figures correspond to the simulation box edges and are a visual artifact of 2D projections with QuickSurf representation. No artificial wall or constraints were introduced laterally. Additional technical details, including the system size and periodic boundary conditions have now been added to the methods section.
It seems quite important that multiple Dyngo molecules aggregate rather than partition into membranes - is this likely to occur in experiment?
Response: Yes, this is important and with the additional simulation experiments suggested by Reviewer #3 it has been clarified that they contribute a great deal to the change in lipid packing of lipid bilayers containing cholesterol. However, it is hard to test aggregation is the cellular system, but we believe that this happens and contribute to the effect on membranes. We have now emphasized the effect of the aggregates in the text.
PMF simulations are strongly suggesting that Dyngo does not spontaneously cross membranes, which is inconsistent with its drug-like amphiphilicity (cLogP~2.5 is optimally suited for membrane permeation) and known effects on intracellular proteins. This suggests an artefact in these PMFs.
Response:As stated in the submitted version of the manuscript, logP was used to validate the topology and the observed value was in a very good agreement with cLogP. Moreover, this validation complemented the standard procedure of CHARMM-GUI ligand modelling, that provided a reasonable penalty score (around 20) for the Dyngo-4a topology. POPC and cholesterol molecules are standard in the force field and validated by numerous studies. The parameters used for the membrane simulations and AWH in particular are very common for this type of studies. Thus, we do not see what may cause any artifacts in the free energy profile construction. In fact, amphiphilicity of the molecule may be one of the key reasons that Dyngo-4a molecule remains at the aqueous interface of the membrane and does not cross the membrane spontaneously. Also, we believe that the energy barrier of 40-60 kJ/mol is not prohibitively high and Dyngo-4a molecules may still overcome the barrier eventually, though we expect majority to reside in the upper leaflet*. *
The authors should experimentally measure the permeation of Dyngo through bilayers (or lack thereof), to more robustly support their finding that Dyngo does not cross membranes spontaneously.
-Response: We thank the reviewer for the suggestion, however this if very technically challenging and would require establishment of precise systems which is beyond the scope of this manuscript.
Why not measure effect of Dyngo on lipid packing directly and more broadly in model membranes?
-Response: With the added modelling experiments supporting the previous simulations and the calculated GP values from the C-Laurdan experiments on cellular plasma membrane, we do not find it necessary to include more model membranes experiments than the already existing ones on lipid monolayers and supported lipid bilayers.
Statistics should not be done on individual cells (n>26), but rather on independent experiment (N=3?)
-Response: We have performed the statistics on live cell particle tracking according to previous literature on similar systems (Boucrot et al., 2011; Larsson et al., 2023; Shvets et al., 2015; Stoeber et al., 2012).
Fig 1G is important but rather unclear. Firstly, these kymographs are an odd way to show that the caveolae are not moving. More importantly, caveolae in normal cells have been shown to be quite stable and immobile (eg doi: 10.1074/jbc.M117.791400), yet here they are claimed to be very mobile.
-Response: Although this might be an odd and unconventional way to depict dynamic processes, we believe that this is a very illustrative way to show track stability over time in bulk rather than just a kymograph over a few structures in a cell. Furthermore, we are not claiming that caveolae are very mobile but rather the opposite very stable in agreement with previous work (Boucrot et al., 2011; Larsson et al., 2023; Mohan et al., 2015). We have now edited the text to make this even clearer.
Also, if Dyngo prevents caveolae scission, there should be more of them at the membrane - why no quantification like Fig 1C to show accumulation of caveolae upon Dyngo treatment? Or directly counting caveolae via EM, as in Fig 4C?
-Response: We are currently preforming CTxB HRP experiments using EM but due to reasons out of our control we have not managed to finish these on time, they will be included in the manuscript once they are ready in hopefully not too long. However, Dynasore has previously been shown, by EM, to increase the number of caveolae at the PM (Moren et al., 2012; Sinha et al., 2011).
The writing can be made more precise and referencing could be strengthened. Response: The introduction was written in a short format, and we have now extended this and made it more precise. Some examples: (a) 'scissoned' is not a word in English,
Response: Thanks, we have now changed this.
(b) what is meant by "Cav1 assembly is driven by high chol content"? There are many types of caveolin assemblies.
Response: We agree that this can be made more precise and have now clarified this in the introduction.
(c) "This generates a unique membrane domain with distinct lipid packing and a very high curvature." Unclear what 'this' refers to and there is no reference here, so what is the evidence for either of these claims? Caveolin-8S oligomers are not curved. Perhaps 'this' is caveolae, but they are relatively large and also not very highly curved and I am unaware of measurements of lipid packing therein.
Response: caveolae are around 50 nm which in biology is a very high curvature of a membrane. It has been extensively proven that caveolae have a distinct lipid composition highly enriched in cholesterol and sphingolipids, which thereby also will generate a unique lipid packing as compared to the surrounding membrane. Yet, the reviewer is correct that lipid packing has not been measured in a caveola for obvious technical challenges. Thus, we have now changed the text to “special lipid composition”.
The sentence following that one again makes a specific, but unreferenced, claim. (d) intro claims that lipid packing is critical for fission, but it is unclear quite what is meant by this claim. The references do not help, as they are often about the basic biophysics of lipids, rather than how packing affects fission.
Response: We have now edited the text.
(e) intro strongly implies that caveolae remain membrane attached because of stalled scission. How strong is the evidence for this? The fact that EHD2 is at the neck is not definitive,
Response: We used the term stalled scission to describe that all omega shaped membrane invaginations do not scission in the same automatic way as clathrin coated vesicles. We have now changed this in the text. Caveolae are shown to be released (undergo scission) and be detected as internal caveolae if the protein EHD2 is removed. Hence this must be interpreted as if EHD2 stalls scission. The evidence includes data compiled over the last 12 years from others and us which include for example: 1) Caveolae with EHD2 have a longer duration time (Larsson et al., 2023; Mohan et al., 2015; Moren et al., 2012; Stoeber et al., 2012), Knock down of EHD2 results in more internalized caveolae as measured by CTxB HRP using EM (Moren et al., 2012) and shorter duration time at the PM (Hubert et al., 2020; Larsson et al., 2023; Mohan et al., 2015; Stoeber et al., 2012). 2) EHD2 overexpression results in less internalized caveolae as measured by CTxB HRP using EM (Stoeber et al., 2012). Furthermore, 3) overexpression or acute addition of purified EHD2 via microinjection counteracts lipid induced scission of caveolae and hence result in caveolae stabilization at the PM (Hubert et al., 2020). It is very hard to see that the release and internalization of caveolae could result from anything else than that these have undergone scission. EHD2 has been found around the rim of caveolae (Matthaeus et al., 2022) and overexpression of EHD2 oligomerizing mutants have been shown to expand the caveola neck (Hoernke et al., 2017; Larsson et al., 2023).
(f) unclear what is meant by 'lipid packing frustration' and how Dyngo supposedly induces it.
Response: Lipid packing frustration refers to what is usually referred to as lipid packing defect, but since lipid membranes are describe as a fluid system it should not have defects whereby, we believe that lipid packing frustration is more accurate. However, we have now changed the text and use “decreased lipid packing” or “decreased lipid order” more thoroughly to describe the effect on the plasma membrane.
IF of Cav1 is insufficient to claim puncta as caveolae. Co-stained puncta of caveolin with cavin are much stronger evidence. Same issue for Cav1-GFP puncta.
Response: We agree and have now provided IF showing cavin1 and EHD2 colocalization to Cav1GFP in non and Dyngo-4a-treated cells.
Fig 3E claims that "preferred position of Dyngo-4a was closer to the head groups" but the minimum looks to be in similar place as Fig 3B without cholesterol.
Response:We appreciate the reviewer’s observation. The PMF minima in the POPC and POPC:Chol membranes are indeed close in absolute position (~1.1–1.2 nm from the bilayer center). However, as clarified in the revised text, the presence of cholesterol leads to a slight shift of Dyngo-4a closer to the headgroup region and broadens the positional distribution. This is also evident from the added density profiles (Fig. S3A) and is now described more precisely in the manuscript.
Critically, these results do not support the notion that Dyngo affects lipid packing sufficiently, which is not measured in the simulations (though could be).
-Response: We thank the reviewer for the excellent suggestion. In response, we have now included a detailed analysis of Dyngo-4a’s effect on lipid packing in the simulations. As described in the revised manuscript, we measured deuterium order parameters, area per lipid (APL), and lipid–Dyngo–cholesterol spatial distributions (Figs. 3-H, S3C-E). The results demonstrate that Dyngo-4a decreases lipid order in POPC:Chol membranes. Both single molecules and clusters reduce the order parameter by up to 0.04 units, particularly in the upper leaflet, where Dyngo-4a reside.The reduction is most pronounced in the midchain region of the sn1 tail and around the double bond of the sn2 tail. These effects were accompanied by increased APL in POPC:Chol membranes and by colocalization of Dyngo-4a near cholesterol-rich regions. Together, these data confirm that Dyngo-4a perturbs membrane organization and lipid packing in a composition-dependent manner. We believe these additions directly address the concern and demonstrate that the simulations indeed support the conclusion that Dyngo-4a modulates lipid packing.
Finally, the simulation data do not show "that Dyngo-4a is competing with cholesterol"; it is unclear what 'competition' means in this context, but regardless, the data only shows that Dyngo sits at a similar location as cholesterol.
We agree with the reviewer that “competition” was an imprecise term. We have rephrased the relevant sections to clarify that Dyngo-4a and cholesterol localize to overlapping regions and exhibit spatial coordination. As now stated in the manuscript, cholesterol appears to partially displace Dyngo-4a from its preferred depth seen in pure POPC, broadens its membrane distribution, and alters lipid packing. According to the order parameters there is an interplay between chol and Dyngo-4a and the heatmaps show that the distribution of chol in the membrane gets less uniform in the presence of Dyngo-4a. These interactions suggest that Dyngo-4a perturbs cholesterol-rich domains.
As new analysis routines were added to the study, we have now also added the details on those to the Methods section of the text.
AFM measures the stiffness of the cell (as correctly explained in Results section) not "overall stiffness of the PM" as stated in the Discussion.
Response: We thank the reviewer for pointing this out, we have now altered this in the discussion section.
Fig2A: what was the starting lipid surface pressure? How does Dyngo insertion depend on initial lipid packing?
Response: The starting pressure lipid pressure was 20 mN m-1 which we now have incorporated in the figure legend. We performed several such experiments with a starting pressure ranging from 20-23 mN m-1 showing consistent results which we described in the materials and methods section. Given that we also performed QCMD analysis and simulations on bilayers showing that Dyngo-4a adsorbed and inserted respectively, we have not performed a titration of starting pressures resulting in a MIP of Dygo-4a.
Fig 4B is a strange approach to measure membrane motion. Why not RMSD or some other displacement based method? As its shown, it implies that the area of the cell changes.
Response: The method that we used to quantify the area of the cell which is attached (or close to) the glass and thereby is visible in TIRF microscopy. This is area indeed changes over time which has been frequently observed and used to describe and quantify the mobility, lamellipodia and filopodia formation among other things. We agree that RMSD can also be used to analyze the data before and after treatments and we have now included RMSD analysis in the manuscript.
Reviewer #3 (Significance (Required)):
The title, abstract, and introduction of the manuscript are largely framed around lipid packing, but most of the data investigate other unexpected effects of treating cells with Dyngo4a. The only measurement for lipid packing (or any other membrane properties) is Fig 4E-F. Therefore, this paper is effectively an investigation of an artefact of a common reagent, which itself could be a valuable contribution. However, the mechanism to explain its effect requires stronger evidence, and its broad biological significance needs further exploration.
Overall, the impact of documenting the effects of Dyngo4a on membranes appears modest but may be valuable to the membrane trafficking community.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
Larsson et al present experimental and computational data on the role of Dyngo4a (a compound that was developed to inhibit dynamin) on the dynamics of caveolae. The manuscript mostly documents effects of Dyngo on caveolae, with one experiment to suggest a mechanism for this result. This one rather unconvincing result forms the focus of the manuscript contributing to a disconnect between the data and the presentation. Additionally, there are concerns with data interpretation. The writing could also benefit from revision to address grammar mistakes, strengthen referencing, and increase precision.
Overall, the manuscript requires substantial revisions before being considered for publication. The central claim, in particular, needs stronger evidence to support the proposed mechanism.
Significant issues (in approximate order of importance):
The title, abstract, and introduction of the manuscript are largely framed around lipid packing, but most of the data investigate other unexpected effects of treating cells with Dyngo4a. The only measurement for lipid packing (or any other membrane properties) is Fig 4E-F. Therefore, this paper is effectively an investigation of an artefact of a common reagent, which itself could be a valuable contribution. However, the mechanism to explain its effect requires stronger evidence, and its broad biological significance needs further exploration.
Overall, the impact of documenting the effects of Dyngo4a on membranes appears modest but may be valuable to the membrane trafficking community.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript uses the small molecule dynamin inhibitors dynasore and dyngo to show that in dynamin triple knockout cells that these inhibitors impact lipid packing and organization in the plasma membrane. Data showing that dyngo affects caveolin dynamics using tirf microscopy is also shown and is interpreted to reflect inhibition of caveolae scission from the membrane.
This data showing that dyngo and dynasore target membrane order is quite compelling and argues that the effects of these inhibitors is not dynamin specific and that inhibition of endocytosis by these small molecule inhibitors is dynamin-independent. The in vitro and in vivo data they provide is convincing.
Similarly, the data showing that dynasore and dyngo affect caveolin dynamics and clathrin endocytosis (transferrin) is quite convincing and argues that altered lipid packing is impacting membrane dynamics at the plasma membrane. What is less convincing is the conclusion is that dyngo is preventing caveolae scission from the membrane. Study of caveolae endocytosis is based on a TIRF assay that has inherent limitations:
My point here is not to discredit the data but only to suggest that the TIRF approach used is an indirect measure of caveolae scission from the membrane that requires substantiation using other approaches.
Dyngo is certainly generally affecting lipid packing via cholesterol and thereby affecting Cav1 dynamics in the plasma membrane. The claim of caveolae scission should be qualified and alternative possibilities considered and discussed. If the authors persist in arguing that dyngo is affecting caveolae scission then the effect should be substantiated by accumulation of caveolae by quantitative EM and high spatial and temporal resolution imaging of Cav1 and cavin1 to define the endocytic events. As the latter represents a new, and potentially very challenging, line of experimentation, I would suggest that it is beyond the scope of the current study. As indicated above the additional experiments are not necessary and qualification of the claims would be sufficient.
Other points
Figure 1C - Cav1 positive spots cannot be interpreted to be caveolae from diffraction limited confocal images. Same comment applies to Fig 4G - caveola? duration.
Figure 4C - it is not clear why this EM data is not quantified - for both the number of caveolae and clathrin coated pits - as this would help clarify the interpretation of the effect reported.
Figure 4D - the AFM experiments should perhaps be repeated as the non-significant effect of dyngo on the Young's modulus may be a result of insufficient n values.
This data showing that dyngo and dynasore target membrane order is quite compelling and argues that the effects of these inhibitors is not dynamin specific and that inhibition of endocytosis by these small molecule inhibitors is dynamin-independent. The in vitro and in vivo data they provide is convincing.
Similarly, the data showing that dynasore and dyngo affect caveolin dynamics and clathrin endocytosis (transferrin) is quite convincing and argues that altered lipid packing is impacting membrane dynamics at the plasma membrane.
What is less convincing is the conclusion is that dyngo is preventing caveolae scission from the membrane.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The authors use Dyngo-4a, a known Dynami inhibitor to test its influence on caveolar assembly and surface mobility. They investigate, whether it incorporates into membranes with Quartz-Crystal Microbalance, they investigate how it is organized in membranes using simulations. Finally, they use lipid-packing sensitive dyes to investigate lipid packing in the presence of Dyngo-4a, membrane stiffness using AFM and membrane undulation using fluorescence microscopy. They also use a measure they call "caveola duration time" to claim that something happens to caveolae after Dyngo-4a addition and using this parameter, they do indeed see an increase in it in response to Dyngo-4a, which is reduced back to the baseline after addition of cholesterol.
Overall, the authors claim: 1) Dyngo-4a inserts into the membrane and this 2) results in "a dramatic dynamin-independent inhibition of caveola scission". 3) Dyngo-4a was inserted and positioned at the level of cholesterol in the bilayer and 4) Dyngo-4a-treatment resulted in decreased lipid packing in the outer leaflet of the plasma membrane 5) but Dyngo-4a did not affect caveola morphology, caveolae- associated proteins, or the overall membrane stiffness 6) acute addition of cholesterol counteracts the block in caveola scission caused by Dyngo-4a
Overall, in this reviewers opinion, claims 1, 3, 4, 5 are well-supported by the presented data from electron and live cell microscopy, QCM-D and AFM. However, there is no convincing assay for caveolar endocytosis presented besides the "caveola duration" which although unclearly described seems to be the time it takes in imaging until a caveolae is not picked up by the tracking software anymore in TIRF microscopy. Since the main claim of the paper is a mechanism of caveolar endocytosis being blocked by Dyngo-4a, a true caveolar internalization assays is required to make this claim. This means either the intracellular detection of not surface connected caveolar cargo or the quantification of caveolar movement from TIRF into epifluorescence detection in the fluorescence microscope. Otherwise, the authors could remove the claim and just claim that caveolar mobility is influenced.
A number of small molecule inhibitors for the GTPase dynamics exist, that are commonly used tools in the investigation of endocytosis. This goes as far that the use of some of these inhibitors alone is considered in some publications as sufficient to declare a process to be dynamin-dependent. However, this is not correct, as there are considerable off-target effects, including the inhibition of caveolar internalization by a dynamin-independent mechanism. This is important, as for example the influence of dynamin small molecule inhibitors on chemotherapy resistance is currently investigated (see for example Tremblay et al., Nature Communications, 2020).
The investigation of the true effect of small molecules discovered as and used as specific inhibitors and their offside effects is extremely important and this reviewer applauds the effort. It is important that inhibitors are not used alone, but other means of targeting a mechanism are exploited as well in functional studies. The audience here thus is besides membrane biophysicists interested in the immediate effect of the small molecule Dyngo-4a also cell biologists and everyone using dynamic inhibitors to investigate cellular function.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
Reviewer comment: This is a very well conceived study of responses to plasma membrane stresses in yeast that signal through the conserved TORC2 complex. Physical stress through small molecular intercalators in the plasma membrane is shown to be independent of their biochemistry and then studies for its effect on plasma membrane morphology and the distribution of free ergosterol (the yeast equivalent of cholesterol), with free being the pool of cholesterol that is available to probes and/or sterol transfer proteins. Experiments nicely demonstrate a negative feedback loop consisting of: stress -> increased free sterol and TORC2 inhibition -> activation of LAM proteins (as demonstrated by Relents and co-workers previously) -> removal of free sterol -> return to unstressed state of PM and TORC2.
Author response: We thank the reviewer for their positive and encouraging feedback. We are pleased to submit our revised manuscript and have addressed all points raised below.
Comment: Fig 2A: Is detection of PIP/PIP2/PS linear for target, or possibly just showing availability that is increased due to local positive curvature?
Response: This is an excellent and fundamental question. While FLARE signal likely reflects lipid availability, its detection is indeed influenced by factors such as membrane curvature and lipid composition, due to varying insertion depths of the lipid-binding domains. For example, studies using NMR suggest that the PLCδ PH domain partially inserts into membranes, potentially conferring curvature sensitivity (Flesch et al., 2005; Uekama et al., 2009). Similarly, curvature influences lactadherin binding, though it's unclear if this extends to its isolated C2 domain (Otzen et al., 2012; Shao et al., 2008; Shi et al., 2004). We could not find direct evidence for curvature sensitivity of P4C(SidC), but assume some influence exists.
To avoid overinterpreting these limitations, we now describe our data based solely on the FLAREs used, rather than inferring enrichment of specific lipid species. We refer to these PM structures as "PI(4,5)P₂-containing", consistent with prior literature (Riggi et al., 2018) and have revised our manuscript accordingly.
Comment: Can any marker be identified for the D4H spots at 2 minutes? In particular, are they early endosomes (shown by brief pre-incubation with FM4-64)?
Response: We appreciate the reviewer's suggestion and have now added new data (Fig. S2E-H). We tested colocalization of D4H spots with FM4-64 (early endosomes), GFP-VPS21 (early endosome marker), and LipidSpot{trade mark, serif} 488 (lipid droplets), but found no overlap. This later observation was not unexpected given that D4H does not recognize Sterol esters. D4H foci also did not overlap with ER (dsRED-HDEL), though they were frequently adjacent to it. While their exact identity remains unknown, we agree this is an intriguing direction for future investigation.
Comment: Is there any functional (& direct) link between Arp inhibition (as in the Pombe study of LAMs by the lab of Sophie Martin) and PM disturbance by amphipathic molecules?
Response: We have explored this connection and now present new data (see final paragraph of Results). Briefly, we show that CK-666 induces internalization of PM sterols in a Lam2/4-dependent manner, and that TORC2 activity is more strongly reduced in lam2Δ lam4Δ cells compared to WT. These findings support the idea that, like PalmC, Arp2/3 inhibition triggers a PM stress that is counteracted by sterol internalization.
Minor Comment: Fig 2A: Labels not clear. Say for each part what FP is used for pip2.
Response: As noted above, we revised image labels to clarify which FLAREs were used, and refer to data accordingly throughout.
Minor Comment: Move fig s2d to main ms. The 1 min and 2 min data are integral to the story.
Response: We agree and have incorporated the 1-min and 2-min data into the main figures. Vehicle-treated controls were moved to Fig. S2.
Minor Comment: The role of Lam2 and Lam4 in retrograde sterol transport has in vivo only been linked to one of their two StART domains not both, as mentioned in the text.
Response: Thank you for pointing this out. We have corrected the text to:
"[...]Lam2 and Lam4[...] contain two START domains, of which at least one has been demonstrated to facilitate sterol transport between membranes (Gatta et al., 2015; Jentsch et al., 2018; Tong et al., 2018)."
Minor Comment: Throughout, images of tagged D4H should be labelled as such, not as "Ergosterol".
Response: We have updated all relevant figure labels and text to refer to "D4H" rather than "Ergosterol", in line with this recommendation.
Reviewer #1 (Significance):
These results in budding yeast are likely to be directly applicable to a wide range of eukaryotic cells, if not all of them. I expect this paper to be a significant guide of research in this area. The paper specifically points out that the current experiments do not distinguish the precise causation among the two outcomes of stress: increased free sterol and TORC2 inhibition. Of these two outcomes which causes which is not yet known. If data were added that shed light on this causation that would make this work much more signifiant, but I can understand 100% that this extra step lies beyond - for a later study for which the current one forms the bedrock.
Response:
We thank the reviewer for their generous assessment. We agree that understanding the causality between increased free sterol and TORC2 inhibition is a critical next step.
Based on our current data, we believe the increase in free ergosterol precedes TORC2 inhibition. For example, TORC2 inhibition alone (e.g., via pharmacological means) does not initially increase free sterol, while it does enhance Lam2/4 activity, promoting sterol internalization (Fig. 3A). Baseline TORC2 activity also inversely correlates with free PM sterol levels in lam2Δ lam4Δ versus LAM2T518A LAM4S401A cells (Figs. 2D, S2C).
Additionally, during sterol depletion, we observe an initial increase in TORC2 activity before growth inhibition occurs, after which activity declines-likely due to compromised PM integrity (Fig. S2M). We now also show that adaptation to several other stresses (e.g., osmotic shock, heat shock, CK-666) partially depends on sterol internalization, which correlates with TORC2 activation (Fig. 4, S4B).
While these findings strengthen the model that PM stress perturbs sterol availability and secondarily impacts TORC2, we cannot yet definitively demonstrate causality. As suggested by Reviewer 3, we tested cholesterol-producing yeast (Souza et al., 2011), but found their response to PalmC indistinguishable from WT, making it difficult to draw mechanistic conclusions (Rebuttal Fig. 2).
Taken together, we favour a model where sterols affect PM properties sensed by TORC2, probably lipid-packing, rather than acting as direct effectors. We hope our revised manuscript more clearly conveys this model and serves as a strong foundation for future mechanistic studies.
Reviewer comment: This manuscript describes multiple effects of positively-charged membrane-intercalating amphipaths (palmitoylcarnitine, PalmC, in particular) on TORC2 in yeast plasma membranes. It is a "next step" in the Loewith laboratory's characterization of the effect of this agent on this system. The study confirms the findings of Riggi et al.(2018) that PalmC inhibits TORC2 and drives the formation of membrane invaginations that contain phosphatidylinositol-bis-phosphate (PIP2) and other anionic phospholipids. It also demonstrates that PalmC intercalates into the membrane, acts directly (rather than through secondary metabolism) and is representative of a class of cationic amphipaths. The interesting finding here is that PalmC causes a rapid initial increase in the plasma membrane ergosterol accessible to the DH4 sterol probe followed by a decrease caused by its transfer to the cytoplasm through its transporter, LAM2/4. TORC2 is implicated in these processes. Loewith et al. have pioneered in this area and this study clearly shows their expertise. Several of the findings reported here are novel. However, I am concerned that PalmC may not be revealing the physiology of the system but rather adding tangential complexity. (This concern applies to the precursor studies using PalmC to probe the TORC2 system.) In particular, I am not confident that the data justify the authors' conclusions "...that TORC2 acts in a feedback loop to control active sterol levels at the PM and [the results] introduce sterols as possible TORC2 signalling modulators."
Author response:
We thank Reviewer #2 for the constructive and critical evaluation of our work. We appreciate the acknowledgment of the novelty and technical strength of several of our findings, and we understand the concern that PalmC could be eliciting non-physiological effects. Our study was designed precisely to use PalmC and similar membrane-active amphipaths as tools to strongly perturb the plasma membrane (PM) in a controlled and tractable way. We now state this intention explicitly in both the Introduction and Discussion sections. To address concerns about the specificity and physiological relevance of PalmC, we have expanded our dataset to include additional PM stressors (hyperosmotic shock, Arp2/3 inhibition, and heat shock), all of which reproduce key features observed with PalmC-namely, TORC2 inhibition, PM invaginations, and retrograde sterol transport (Fig. 4, S4).
We hope this more comprehensive dataset, along with revised discussion and clarified claims, addresses the reviewer's concerns regarding physiological interpretation and artifact.
Major issues 1 and 2: 1. The invaginations induced by PalmC may not be physiologic but simply the result of the well-known "bilayer couple" bending of the bilayer due to the accumulation of cationic amphipaths in the inner leaflet of the plasma membrane bilayer which is rich in anionic phospholipids. Such unphysiological effects make the observed correlation of invagination with TORC2 inhibition etc. hard to interpret.
Electrostatic/hydrophobic association of PIP2 with PalmC could sequester the anionic phospholipid(s). Such associations could also drive the accumulation of PIP2 in the invaginations. This could explain PalmC inhibition of TORC2 through a simple physical rather than biological process. So, it is difficult to draw any physiological conclusion about PIP2 from these experiments.
Response to major issues 1 and 2:
We agree that amphipath-induced bilayer stress, including via the bilayer-couple mechanism, may contribute to PM curvature changes. However, the reviewer's assumption that PalmC inserts preferentially into the inner leaflet appears inconsistent with both literature and our observations. PalmC is zwitterionic, not cationic, and is unlikely to electrostatically sequester anionic lipids such as PIP2. For clarification, we included a short summary of our proposed mechanism of PalmC in the context of the current literature in our Discussion:
"[...] study it was also demonstrated that addition of phospholipids to the outer PM leaflet causes an excess of free sterol at the inner PM leaflet, and its subsequent retrograde transport to lipid droplets (Doktorova et al., 2025). Although we cannot exclude that it is the substrate of a flippase or scramblase, PalmC is not a metabolite found in yeast, nor, given its charged headgroup, is it likely to spontaneously flip to the inner leaflet (Goñi, Requero and Alonso, 1996). Thus, we propose that PalmC accumulates in the outer leaflet, disrupts the lipid balance with the inner leaflet which is, similarly to the mammalian cell model (Doktorova et al., 2025), rectified by sterol mobilization, flipping and internalization (Fig. 5B)."
While we agree that PM invaginations per se are not the central focus of this study, they are indeed a reproducible and biologically intriguing phenomenon. We emphasize that similar invaginations occur not only during PalmC treatment but also in response to other physiological stresses, such as hyperosmotic shock and Arp2/3 inhibition (Fig. 4), and have been reported independently by others (Phan et al., 2025). Furthermore, related structures have been documented in yeast mutants with altered PIP2 metabolism or TORC2 hyperactivity (Rodríguez-Escudero et al., 2018; Sakata et al., 2022; Stefan et al., 2002), and even in mammalian neurons with SJ1 phosphatase mutations (Stefan et al., 2002). These observations support our interpretation that the observed invaginations represent an exaggerated manifestation of a physiologically relevant stress-adaptive process. In our previous study we indeed proposed that PI(4,5)P2 enrichment in PM invaginations was important for PalmC-induced TORC2 inactivation, using the heat sensitive PI(4,5)P2 kinase allele mss4ts - a rather blunt tool (Riggi et al., 2018). We have now come to the conclusion that different mechanisms other than, or in addition to, PIP2 changes drive TORC2 inhibition in our system. In this study, we use the 2xPH(PLC) FLARE exclusively as a generic PM marker, not as a readout of PIP2 biology. Rather, we propose that sterol redistribution and/or the biophysical impact that this has on the PM are central drivers, with TORC2 acting as a signaling node that senses and adjusts PM composition accordingly.
We now clarify these arguments in the revised Discussion and have reframed our use of PalmC as a probe to explore the capacity of the PM to adapt to acute stress via dynamic lipid rearrangements.
Major issue 3:
As the authors point out, a large number of intercalated amphipaths displace sterols from their association with bilayer phospholipids. This unphysiologic mechanism can explain how PalmC causes the transient increase in the availability of plasma membrane ergosterol to the D4H probe and its subsequent removal from the plasma membrane via LAM2/4. TORC2 regulation may not be involved. In fact, the authors say that "TORC2 inhibition, and thereby Lam2/4 activation, cannot be the only trigger for PalmC induced sterol removal." Furthermore, the subsequent recovery of plasma membrane ergosterol could simply reflect homeostatic responses independent of the components studied here.
Response:
We agree that increased free sterols in the inner leaflet likely initiate retrograde transport. Our results suggest that TORC2 inhibition facilitates this process by disinhibiting Lam2/4, allowing more efficient clearance of ergosterol from the PM (Fig. 3A, S2C). However, the process is not exclusively dependent on TORC2, and we state this explicitly.
We do not observe recovery of PM ergosterol on the timescales measured, while TORC2 activity recovers, suggesting that restoration likely occurs later via biosynthetic or anterograde trafficking pathways, which are outside the scope of this study. These points are clarified in the revised Discussion.
Major issue 3a:
The data suggest that LAM2/4 mediates the return of cytoplasmic ergosterol to the plasma membrane. To my knowledge, this is a nice finding that not been reported previously and is worth confirming more directly.
Response:
We thank the reviewer for this observation but would like to clarify a misunderstanding: our data do not suggest that Lam2/4 mediates anterograde sterol transport. Our results and prior work (Gatta et al., 2015; Roelants et al., 2018) show that Lam2/4 mediate retrograde transport from the PM to the ER, and TORC2 inhibits this process. We now clarify this point in the revised manuscript, stating:
"In vivo, Lam2/4 seem to predominantly transport sterols from the PM to the ER, following the concentration gradient (Gatta et al., 2015; Jentsch et al., 2018; Tong et al., 2018)."
Major issue 4:
I agree with the authors that "It is unclear if the excess of free sterols itself is part of the inhibitory signal to TORC2..." Instead, the inhibition of TORC2 by PalmC may simply result from its artifactual aggregation of the anionic phospholipids (especially, PIP2) needed for TORC2 activity. This would not be biologically meaningful. If the authors wish to show that accessible ergosterol inhibits TORC2 activity or vice versa, they should use more direct methods. For example, neutral amphipaths that do not cause the aforementioned PalmC perturbations should still increase plasma membrane ergosterol and send it through LAM2/4 to the ER.
Response:
We now provide evidence that three orthologous treatments (hyperosmotic shock, heat shock and Arp2/3 inhibition) similarly cause sterol mobilization and, in the absence of sterol clearance from the PM, prolonged TORC2 inhibition. These results do not support the reviewer's contention that the inhibition of TORC2 by PalmC is simply resulting from its artifactual aggregation of the anionic phospholipids. Furthermore, PalmC is zwitterionic, and its interaction with anionic lipids should be somewhat limited.
In our experimental setup, neutral amphipaths did not trigger TORC2 inhibition or D4H redistribution While this differs from prior in vitro work (Lange et al., 2009), we attribute this in part to a discrepancy to experimental setup differences, including flow chamber artifacts that we discuss in the methods section.
Importantly, only amphipaths with a charged headgroup, including zwitterionic (PalmC) and positively charged analogs, produced robust effects. A negatively charged derivative also seemed to have a minor effect on TORC2 activity and PM sterol internalization (Palmitoylglycine (Fig. 1D, Rebuttal Fig. 1). This suggests that in vivo, charge-based membrane perturbation is required to alter PM sterol distribution and TORC2 activity.
Major issue 5.:
The mechanistic relationship between TORC2 activity and ergosterol suggested in the title, abstract, and discussion is not secure. I agree with the concluding section of the manuscript called "Limitations of the study". It highlights the need for a better approach to the interplay between TORC2 and ergosterol.
Response:
This may have been true of the previous submission, but we now demonstrate that provoking PM stress in four orthogonal ways triggers mobilization of sterols, which left uncleared, prevents normal (re)activation of TORC2 activity. We thus conclude that free sterols, directly or more likely indirectly, inhibit TORC2. The role that TORC2 plays in sterol retrotranslocation has been demonstrated previously (Roelants et al., 2018). We believe our expanded data and clarified framework make a compelling case for a stress-adaptive role of sterol retrograde transport that is supervised and modulated-but not fully driven-by TORC2 activity.
Thus, we feel in the present version of this manuscript that the title is now justified.
Minor issue: Based on earlier work using the reporter fliptR, the authors claim that PalmC reduces membrane tension. They should consider that this intercalated dye senses many variables including membrane tension but also lipid packing. I suspect that, by intercalating into and thereby altering the bilayer, PalmC is affecting the latter rather than the former.
Response:
We thank the reviewer for this important point regarding the multifactorial sensitivity of intercalating dyes such as Flipper-TR®, including to membrane tension and lipid packing.
We respectfully note, however, that our current study does not include any new data generated using Flipper-TR®. We referred to earlier work (Riggi et al., 2018) for context, where Flipper-TR® was used as a membrane tension reporter.
We fully agree that the response of such "smart" membrane probes integrates multiple biophysical parameters-including tension, packing, and hydration-which are themselves interrelated as consequences of membrane composition (Colom et al., 2018; Ragaller et al., 2024; Torra et al., 2024). Indeed, this interconnectedness is central to our interpretation of PalmC's pleiotropic effects on the plasma membrane (PM). In our previous study, we observed that PalmC treatment not only reduced apparent PM tension (as measured by Flipper-TR®) but also increased membrane order ((Riggi et al., 2018); see laurdan GP, Fig. 6C), and here we show that it promotes the redistribution of free sterol away from the PM.
Furthermore, PalmC's effect on membrane tension was supported by orthogonal in vitro data: its addition to giant unilamellar vesicles (GUVs) led to a measurable increase in membrane surface area and decreased tension, as shown by pipette aspiration ((Riggi et al., 2018), Fig. 3F). This provides complementary evidence that the membrane tension reduction is not merely an artifact of Flipper-TR® reporting.
That said, we agree with the reviewer that in the case of TORC2 inhibition or hyperactivation, the observed changes in PM tension are based solely on Flipper-TR® data, without additional orthogonal validation. To address this concern, we have revised the relevant text in the manuscript to more cautiously reflect this complexity. The revised sentence now reads:
"Consistent with this role, data generated with the lipid packing reporter dye Flipper-TR® suggest that acute chemical inhibition of TORC2 increases PM tension, while Ypk1 hyperactivation decreases it."
This revised phrasing acknowledges both the utility and the limitations of Flipper-TR® as a probe of membrane biophysics.
Reviewer #2 Significance:
This is an interesting topic. However, use of the exogenous probe, palmitoylcarnitine, could be causing multiple changes that complicate the interpretation of the data.
Reviewers #1 and #3 were much more impressed by this study than I was. I am not a yeast expert and so I may have missed or confused something. I would therefore welcome their expert feedback regarding my comments (#2). Ted Steck
Response:
Thank you for your constructive feedback.
We believe that the manuscript is now much improved, and we hope to have convinced you that the mechanisms that we've elucidated using PalmC represent a general adaptation response to physiological PM stressors.
Reviewer comment: The authors describe the effects of surfactant-like molecules on the plasma membrane (PM) and its associated TORC2 complex. Addition of the surfactants with a positively-charged headgroup and a hydro-carbon tail of at least 16 caused the rapid clustering of PI-4,5P2 together with PI-4P and phosphatidylserine in large membrane invaginations. The authors convincingly demonstrate that this effect of the surfactants on the PM is likely caused by a direct disturbance of the PM organization and/or lipid composition. Interestingly, upon PalmC treatment, free ergosterol of the PM was found to first concentrate in the clusters, but within The kinetics of the changes in free ergosterol levels and the changes in TORC2 activity do not match. Ergosterol is rapidly depleted after PalmC treatment (The Lam2/4 data support the idea that ergosterol transport plays a role in the TORC2 recovery, but what role this is, is not clear to me. I think the data fit better with a model in which PalmC causes low tension of the PM which in turn disrupts normal lipid organization and thus causes TORC2 to shut down, maybe not by changes in free ergosterol but by changes, for instance, in lipid raft formation (which is in part effected by ergosterol levels). The transport of ergosterol is only one mechanism that is involved in restoring PM tension and TORC2 activity. However, sensing free ergosterol alone is most likely not the mechanism explaining how TORC2 senses PM tension.
Therefore, I recommend that the model is revised (or supported by more data), reflecting the fact that free ergosterol levels do not directly correlate with the TORC2 activity, but instead might be only one of the PM parameters that regulate TORC2.
Author response:
We thank the reviewer for their thoughtful assessment and constructive suggestions. As described in more detail above, we have included in our revised version of this manuscript a variety of new data, including the sterol-internalization dependent adaptation of the PM and regulation of TORC2 during additional stresses. We think that these data vastly improve on our previous manuscript version. We have addressed each point risen by the reviewer below and revised the manuscript accordingly, including a rewritten discussion and updated model to better reflect the limitations of our current understanding of how TORC2 senses changes in the plasma membrane (PM). It is true that the appearance of PM invaginations tracks well with TORC2 inhibition, but it is not clear to us if they are upstream of this inhibition or merely another symptom of the preceding PM perturbation (PalmC-induced free sterol increase can be observed after 10s (Fig. S2A), but PM invaginations become visible only after ~1 min - meanwhile we can observe near complete TORC2 inhibition after 30s). In this study, we are mostly interested in the role of PM sterol redistribution in stress response. Indeed we think that the role of free sterol clearance during stresses is to adapt the PM to these stresses - thus restoring PM parameters which in turn reactivates TORC2. This can be seen for hyperosmotic stress and the newly introduced PM stressors, Arp2/3 inhibition and heat shock response (Fig. 4). We have therefore softened our model and updated discussion and final figure (Fig. 5) to reflect that TORC2 likely responds to broader changes in PM organization or tension, with sterol redistribution representing one of several contributing factors rather than the sole signal.
Comment: - If TORC2 is indeed inhibited by free ergosterol, the addition of ergosterol to the growth medium should be able to trigger similar effects as PalmC. If this detection of free ergosterol is very specific (e.g. if TORC2 has a binding pocket for ergosterol) we would expect that addition of other sterols such a cholesterol or ergosterol precursors should not inhibit TORC2.
Response:
We appreciate this suggestion and agree that testing whether exogenous ergosterol can mimic PalmC effects would help assess specificity. However, yeast do not readily take up sterols under aerobic conditions, which renders artificial sterol enrichment at the yeast PM rather difficult. We have now included additional data characterizing our Lam2/4 mutants (see below), and pharmacological sterol synthesis inhibition, showing that a depletion of free sterols from the PM correlates with lower TORC2 activity (Fig. 2D, S2C). Additionally, as suggested, we tried to probe if ergosterol directly interacts with TORC2 through a specific binding pocket, by treating a yeast strain expressing cholesterol rather than ergosterol (Souza et al., 2011) with PalmC. However, the response of TORC2 activity in these cells was very similar to that of WT cells (Rebuttal Fig. 2). In conclusion, we agree that at present we do not know mechanistically how sterols affect TORC2 activity, although it does indeed seem more likely to be through an indirect mechanism linked to changes in PM parameters. The nature of such a mechanism will be subject to further studies. We hope that the introduced changes to the manuscript adequately reflect these considerations.
Rebuttal Fig. 2: WT yeast cells which produce ergosterol as main sterol, and mutant cells which produce cholesterol instead were treated with 5 µM PalmC, and TORC2 activity was assessed by relative phosphorylation of Ypk1 on WB. One representative experiment out of two replicates.
Comment: - The experiment in Figure 1C is not controlled for differences in membrane intercalation of the different compounds. For instance, does C16 choline and C16 glycine accumulate at the same rate in the PM (measure similar to experiment in Figure 1B). Maybe the positive charge at the headgroup of the surfactants increases the local concentration at the PM and therefore can explain the difference in effect on the PM.
Response:
We agree with the reviewer that the effects of the various PalmC derivatives are not directly controlled for differences in membrane intercalation. Our structure-activity screen was intended to demonstrate the general biophysical mode of action of PalmC-like compounds and to define minimal structural requirements for activity.
We now note in the manuscript that differential membrane insertion could contribute to the observed variation in efficacy, particularly in relation to tail length. While we considered this additional suggested experiment, it was ultimately judged to be outside the scope of this study due to its complexity and limited impact on the central conclusions.
A clarifying sentence has been added to the relevant results section to explicitly acknowledge this limitation:
"We did not control for differences in PM intercalation efficiency."
We also include a discussion here to further clarify our interpretation. Prior in vitro studies have shown that while intercalation is necessary, it is not sufficient for PM perturbation. For example, palmitoyl-CoA intercalates into membranes but does not induce the same biophysical effects as PalmC (Goñi et al., 1996; Ho et al., 2002). Thus, we believe that intercalation is only part of the story, and that the intrinsic propensity of different headgroups to perturb the PM plays a key role in the disruption of PM lipid organization.
Comment: - Are the intracellular ergosterol structures associated (or in close proximity) with lipid droplets (ergosterol being modified and delivered into a lipid droplet)?
Response:
We thank the reviewer for raising this point. We now include additional data (Fig. S2H) showing that intracellular D4H-positive structures do not reside near or colocalize with lipid droplets. The latter is not entirely unexpected as D4H does not recognize esterified sterols. However, we do observe an increase in overall LD volume following PalmC treatment, consistent with the idea that internalized PM sterols may be stored in LDs as sterol esters over time - although we did not test if this increase in LD volume is Lam2/4 dependent. This increase is mentioned in the revised results text. An increase in cellular LDs has also been recently reported during hyperosmotic shock (Phan et al., 2025).
For more attempts to identify a marker for intracellular D4H foci, see reply to reviewer 1.
Comment:
Response:
We thank the reviewer for this question, as in the course of generating these data we realized that our "inhibited" DD mutant was in fact not phosphomimetic but displayed the same D4H distribution as the "hyperactive" AA mutant, i.e. a marked inwards shift of D4H signal away from the PM to internal structures due to increased PM-ER retrograde transport of sterols (Fig. S2C). This led us to critically re-evaluate and ultimately repeat our TORC2 activity WB experiments for PalmC treatment in LAM2/4 mutants. In this new set of experiments, the faster TORC2 recovery after PalmC treatment in the LAM2T518A LAM4S401A mutant did unfortunately not repeat robustly. It is possible that such differences can be observed under specific conditions. Nevertheless, the improved overall quality of the Western blot data allowed us to make the observation that baseline activity was already slightly different in these strains. The Lam2/4 centered part of the results section has subsequently been updated in the manuscript:
"Using a phosphospecific antibody, we did not observe an increase in baseline TORC2 activity in lam2Δ lam4Δ cells, which had been previously reported by electrophoretic mobility shift (Murley et al., 2017). Instead, baseline TORC2 activity was consistently slightly decreased in these cells (Fig. 2D). Ypk1, activated directly by TORC2, inhibits Lam2 and Lam4 through phosphorylation on Thr518 and Ser401, respectively (Roelants et al., 2018; Topolska et al., 2020). We substituted these residues with alanine, generating a strain in which Lam2/4 were no longer inhibited by phosphorylation (Roelants et al., 2018). In these cells, yeGFP-D4H showed that free sterols were constitutively shifted away from the PM to intracellular structures (Fig. S2C, bottom panel). Intriguingly, in opposition to lam2Δ lam4Δ cells, basal TORC2 activity was increased in LAM2T518A LAM4S401A cells (Fig. 2D). This suggests that a decrease in free PM sterols stimulates TORC2 activity [...]"
"In LAM2T518A LAM4S401A cells, TORC2 activity recovers with similar kinetics as the WT (Fig. 2D, bottom blot), suggesting that Lam2/4 release from TORC2 dependent inhibition during PalmC treatment is a fast and efficient process in WT cells, not further expedited by these constitutively active Lams."
As suggested, we also observed D4H localization in LAM2T518A LAM4S401A after PalmC treatment, and implemented these data to further demonstrate that PalmC causes an increase in the fraction of free ergosterol at the PM, which is subsequently removed:
"PalmC addition to LAM2T518A LAM4S401A cells likewise resulted first in a transient increase and then a further decrease in PM yeGFP-D4H signal (Fig. 3C, S3D)."
Comment: - Does Lam2/4 localize to ER-PM contact sites near the large PM invaginations, which could allow for efficient transport of the free ergosterol that accumulates in these structures.
Response:
We were curious about this too, and have now added the requested data in our supplementary material and added a sentence in our results:
"Indeed, in cells expressing GFP-Lam2 we observed that PalmC induced PM invaginations often formed at sites with preexisting GFP-Lam2 foci (Fig. S2K, cyan arrow), although GFP-Lam2 foci did not always colocalize with invaginations (Fig. S2K, yellow arrow) and vice versa. "
Additionally, in the effort to characterize intracellular D4H foci during PalmC as requested by reviewer 1, we also looked at the localization of these foci relative to ER, and found that
"During early timepoints, intracellular foci are usually in close vicinity to ER (Fig. S2E)"
Reviewer #3 (Significance (Required)): The manuscript describes the effects of small molecule surfactants on the PM organization and on TORC2 activity. This is an important set of observation that helps understanding the response of cells to environmental stressors that affect the PM. This field of study is very challenging because of the limited tools available to directly observe lipids and their movements. I consider the data and most of its interpretations of high importance, but I am not convinced of the larger model that tries to link the ergosterol data with TORC2 activity. With adjustments of the model or additional experimental support, this manuscript will be of general interest for cell biologists, especially for researchers studying membrane stress response pathways.
Response:
We thank the reviewer for highlighting the importance of studying PM stress responses and acknowledging the technical challenges involved. We hope the applied changes and additional data succeed in softening our claims about TORC2 regulation while convincing the reviewer that free sterol levels at the PM are one of several contributing factors that correlate with changes in TORC2 activity.
Colom, A., Derivery, E., Soleimanpour, S., Tomba, C., Dal Molin, M., Sakai, N., González-Gaitán, M., Matile, S., Roux, A., 2018. A Fluorescent Membrane Tension Probe. Nat. Chem. 10, 1118-1125. https://doi.org/10.1038/s41557-018-0127-3
Flesch, F.M., Yu, J.W., Lemmon, M.A., Burger, K.N.J., 2005. Membrane activity of the phospholipase C-δ1 pleckstrin homology (PH) domain. Biochem. J. 389, 435-441. https://doi.org/10.1042/BJ20041721
Gatta, A.T., Wong, L.H., Sere, Y.Y., Calderón-Noreña, D.M., Cockcroft, S., Menon, A.K., Levine, T.P., 2015. A new family of StART domain proteins at membrane contact sites has a role in ER-PM sterol transport. eLife 4. https://doi.org/10.7554/eLife.07253
Goñi, F.M., Requero, M.A., Alonso, A., 1996. Palmitoylcarnitine, a surface-active metabolite. FEBS Lett. 390, 1-5. https://doi.org/10.1016/0014-5793(96)00603-5
Ho, J.K., Duclos, R.I., Hamilton, J.A., 2002. Interactions of acyl carnitines with model membranes: a (13)C-NMR study. J. Lipid Res. 43, 1429-1439. https://doi.org/10.1194/jlr.m200137-jlr200
Jentsch, J.-A., Kiburu, I., Pandey, K., Timme, M., Ramlall, T., Levkau, B., Wu, J., Eliezer, D., Boudker, O., Menon, A.K., 2018. Structural basis of sterol binding and transport by a yeast StARkin domain. J. Biol. Chem. 293, 5522-5531. https://doi.org/10.1074/jbc.RA118.001881
Murley, A., Yamada, J., Niles, B.J., Toulmay, A., Prinz, W.A., Powers, T., Nunnari, J., 2017. Sterol transporters at membrane contact sites regulate TORC1 and TORC2 signaling. J. Cell Biol. 216, 2679-2689. https://doi.org/10.1083/jcb.201610032
Otzen, D.E., Blans, K., Wang, H., Gilbert, G.E., Rasmussen, J.T., 2012. Lactadherin binds to phosphatidylserine-containing vesicles in a two-step mechanism sensitive to vesicle size and composition. Biochim. Biophys. Acta BBA - Biomembr., Protein Folding in Membranes 1818, 1019-1027. https://doi.org/10.1016/j.bbamem.2011.08.032
Phan, J., Silva, M., Kohlmeyer, R., Ruethemann, R., Gay, L., Jorgensen, E., Babst, M., 2025. Recovery of plasma membrane tension after a hyperosmotic shock. Mol. Biol. Cell 36, ar45. https://doi.org/10.1091/mbc.E24-10-0436
Ragaller, F., Sjule, E., Urem, Y.B., Schlegel, J., El, R., Urbancic, D., Urbancic, I., Blom, H., Sezgin, E., 2024. Quantifying Fluorescence Lifetime Responsiveness of Environment-Sensitive Probes for Membrane Fluidity Measurements. J. Phys. Chem. B 128, 2154-2167. https://doi.org/10.1021/acs.jpcb.3c07006
Riggi, M., Niewola-Staszkowska, K., Chiaruttini, N., Colom, A., Kusmider, B., Mercier, V., Soleimanpour, S., Stahl, M., Matile, S., Roux, A., Loewith, R., 2018. Decrease in plasma membrane tension triggers PtdIns(4,5)P2 phase separation to inactivate TORC2. Nat. Cell Biol. 20, 1043-1051. https://doi.org/10.1038/s41556-018-0150-z
Rodríguez-Escudero, I., Fernández-Acero, T., Cid, V.J., Molina, M., 2018. Heterologous mammalian Akt disrupts plasma membrane homeostasis by taking over TORC2 signaling in Saccharomyces cerevisiae. Sci. Rep. 8, 7732. https://doi.org/10.1038/s41598-018-25717-w
Roelants, F.M., Chauhan, N., Muir, A., Davis, J.C., Menon, A.K., Levine, T.P., Thorner, J., 2018. TOR complex 2-regulated protein kinase Ypk1 controls sterol distribution by inhibiting StARkin domain-containing proteins located at plasma membrane-endoplasmic reticulum contact sites. Mol. Biol. Cell 29, 2128-2136. https://doi.org/10.1091/mbc.E18-04-0229
Sakata, K.-T., Hashii, K., Yoshizawa, K., Tahara, Y.O., Yae, K., Tsuda, R., Tanaka, N., Maeda, T., Miyata, M., Tabuchi, M., 2022. Coordinated regulation of TORC2 signaling by MCC/eisosome-associated proteins, Pil1 and tetraspan membrane proteins during the stress response. Mol. Microbiol. 117, 1227-1244. https://doi.org/10.1111/mmi.14903
Shao, C., Novakovic, V.A., Head, J.F., Seaton, B.A., Gilbert, G.E., 2008. Crystal Structure of Lactadherin C2 Domain at 1.7Å Resolution with Mutational and Computational Analyses of Its Membrane-binding Motif*. J. Biol. Chem. 283, 7230-7241. https://doi.org/10.1074/jbc.M705195200
Shi, J., Heegaard, C.W., Rasmussen, J.T., Gilbert, G.E., 2004. Lactadherin binds selectively to membranes containing phosphatidyl-L-serine and increased curvature. Biochim. Biophys. Acta 1667, 82-90. https://doi.org/10.1016/j.bbamem.2004.09.006
Souza, C.M., Schwabe, T.M.E., Pichler, H., Ploier, B., Leitner, E., Guan, X.L., Wenk, M.R., Riezman, I., Riezman, H., 2011. A stable yeast strain efficiently producing cholesterol instead of ergosterol is functional for tryptophan uptake, but not weak organic acid resistance. Metab. Eng. 13, 555-569. https://doi.org/10.1016/j.ymben.2011.06.006
Stefan, C.J., Audhya, A., Emr, S.D., 2002. The yeast synaptojanin-like proteins control the cellular distribution of phosphatidylinositol (4,5)-bisphosphate. Mol. Biol. Cell 13, 542-557. https://doi.org/10.1091/mbc.01-10-0476
Tong, J., Manik, M.K., Im, Y.J., 2018. Structural basis of sterol recognition and nonvesicular transport by lipid transfer proteins anchored at membrane contact sites. Proc. Natl. Acad. Sci. 115, E856-E865. https://doi.org/10.1073/pnas.1719709115
Topolska, M., Roelants, F.M., Si, E.P., Thorner, J., 2020. TORC2-Dependent Ypk1-Mediated Phosphorylation of Lam2/Ltc4 Disrupts Its Association with the β-Propeller Protein Laf1 at Endoplasmic Reticulum-Plasma Membrane Contact Sites in the Yeast Saccharomyces cerevisiae. Biomolecules 10, 1598. https://doi.org/10.3390/biom10121598
Torra, J., Campelo, F., Garcia-Parajo, M.F., 2024. Tensing Flipper: Photosensitized Manipulation of Membrane Tension, Lipid Phase Separation, and Raft Protein Sorting in Biological Membranes. J. Am. Chem. Soc. 146, 24114-24124. https://doi.org/10.1021/jacs.4c08580
Uekama, N., Aoki, T., Maruoka, T., Kurisu, S., Hatakeyama, A., Yamaguchi, S., Okada, M., Yagisawa, H., Nishimura, K., Tuzi, S., 2009. Influence of membrane curvature on the structure of the membrane-associated pleckstrin homology domain of phospholipase C-δ1. Biochim. Biophys. Acta BBA - Biomembr. 1788, 2575-2583. https://doi.org/10.1016/j.bbamem.2009.10.009
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
The authors describe the effects of surfactant-like molecules on the plasma membrane (PM) and its associated TORC2 complex. Addition of the surfactants with a positively-charged headgroup and a hydro-carbon tail of at least 16 caused the rapid clustering of PI-4,5P2 together with PI-4P and phosphatidylserine in large membrane invaginations. The authors convincingly demonstrate that this effect of the surfactants on the PM is likely caused by a direct disturbance of the PM organization and/or lipid composition. Interestingly, upon PalmC treatment, free ergosterol of the PM was found to first concentrate in the clusters, but within <5min this ergosterol seemed to be transported into intracellular structures, causing an overall loss in free ergosterol of the PM. The authors speculate that the initial spike in free ergosterol might be the trigger for the shutdown of TORC2 signaling. The PalmC-triggered transport of free ergosterol from the PM to intracellular structures required the lipid transport proteins Lam2/4. Loss of these transporters caused a delay in TORC2 reactivation, supporting the idea that ergosterol transport out of the PM plays a role in the recovery of normal PM organization. Hyperosmotic shock mimics some of the effects observed with PamlC, but unlike PalmC treatment, TORC2 recovery after hyperosmotic shock is not dependent on Lam2/4.
The presented data are of high quality and most conclusions are well supported. However, based on the presented data the model that a PalmC-triggered increase in free ergosterol is the cause of TORC2 inactivation is not obvious to me. The kinetics of the changes in free ergosterol levels and the changes in TORC2 activity do not match. Ergosterol is rapidly depleted after PalmC treatment (<5min) whereas TORC2 activity requires 30min to recover. Also, the hyperosmotic data on free ergosterol levels and TORC2 activity do not match. In fact, the presence of the large PM invaginations is a better predictor of TORC2 activity. The Lam2/4 data support the idea that ergosterol transport plays a role in the TORC2 recovery, but what role this is, is not clear to me. I think the data fit better with a model in which PalmC causes low tension of the PM which in turn disrupts normal lipid organization and thus causes TORC2 to shut down, maybe not by changes in free ergosterol but by changes, for instance, in lipid raft formation (which is in part effected by ergosterol levels). The transport of ergosterol is only one mechanism that is involved in restoring PM tension and TORC2 activity. However, sensing free ergosterol alone is most likely not the mechanism explaining how TORC2 senses PM tension. Therefore, I recommend that the model is revised (or supported by more data), reflecting the fact that free ergosterol levels do not directly correlate with the TORC2 activity, but instead might be only one of the PM parameters that regulate TORC2.
Further comments:
The manuscript describes the effects of small molecule surfactants on the PM organization and on TORC2 activity. This is an important set of observation that helps understanding the response of cells to environmental stressors that affect the PM. This field of study is very challenging because of the limited tools available to directly observe lipids and their movements. I consider the data and most of its interpretations of high importance, but I am not convinced of the larger model that tries to link the ergosterol data with TORC2 activity. With adjustments of the model or additional experimental support, this manuscript will be of general interest for cell biologists, especially for researchers studying membrane stress response pathways.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This manuscript describes multiple effects of positively-charged membrane-intercalating amphipaths (palmitoylcarnitine, PalmC, in particular) on TORC2 in yeast plasma membranes. It is a "next step" in the Loewith laboratory's characterization of the effect of this agent on this system. The study confirms the findings of Riggi et al.(2018) that PalmC inhibits TORC2 and drives the formation of membrane invaginations that contain phosphatidylinositol-bis-phosphate (PIP2) and other anionic phospholipids. It also demonstrates that PalmC intercalates into the membrane, acts directly (rather than through secondary metabolism) and is representative of a class of cationic amphipaths. The interesting finding here is that PalmC causes a rapid initial increase in the plasma membrane ergosterol accessible to the DH4 sterol probe followed by a decrease caused by its transfer to the cytoplasm through its transporter, LAM2/4. TORC2 is implicated in these processes.
Loewith et al. have pioneered in this area and this study clearly shows their expertise. Several of the findings reported here are novel. However, I am concerned that PalmC may not be revealing the physiology of the system but rather adding tangential complexity. (This concern applies to the precursor studies using PalmC to probe the TORC2 system.) In particular, I am not confident that the data justify the authors' conclusions "...that TORC2 acts in a feedback loop to control active sterol levels at the PM and [the results] introduce sterols as possible TORC2 signalling modulators."
Major issues
3a. The data suggest that LAM2/4 mediates the return of cytoplasmic ergosterol to the plasma membrane. To my knowledge, this is a nice finding that not been reported previously and is worth confirming more directly. 4. I agree with the authors that "It is unclear if the excess of free sterols itself is part of the inhibitory signal to TORC2..." Instead, the inhibition of TORC2 by PalmC may simply result from its artifactual aggregation of the anionic phospholipids (especially, PIP2) needed for TORC2 activity. This would not be biologically meaningful. If the authors wish to show that accessible ergosterol inhibits TORC2 activity or vice versa, they should use more direct methods. For example, neutral amphipaths that do not cause the aforementioned PalmC perturbations should still increase plasma membrane ergosterol and send it through LAM2/4 to the ER. 5. The mechanistic relationship between TORC2 activity and ergosterol suggested in the the title, abstract and discussion is not secure. I agree with the concluding section of the manuscript called "Limitations of the study". It highlights the need for a better approach to the interplay between TORC2 and ergosterol.
Minor issue
Based on earlier work using the reporter fliptR, the authors claim that PalmC reduces membrane tension. They should consider that this intercalated dye senses many variables including membrane tension but also lipid packing. I suspect that, by intercalating into and thereby altering the bilayer, PalmC is affecting the latter rather than the former.
Referees cross-commenting
Reviewers #1 and #3 were much more impressed by this study than I was. I am not a yeast expert and so I may have missed or confused something. I would therefore welcome their expert feedback regarding my comments (#2). Ted Steck
This is an interesting topic. However, use of the exogenous probe, palmitoylcarnitine, could be causing multiple changes that complicate the interpretation of the data.
Note: This preprint has been reviewed by subject experts for Review Commons. Content has not been altered except for formatting.
Learn more at Review Commons
This is a very well conceived study of responses to plasma membrane stresses in yeast that signal through the conserved TORC2 complex.
Physical stress through small molecular intercalators in the plasma membrane is shown to be independent of their biochemistry and then studies for its effect on plasma membrane morphology and the distribution of free ergosterol (the yeast equivalent of cholesterol), with free being the pool of cholesterol that is available to probes and/or sterol transfer proteins. Experiments nicely demonstrate a negative feedback loop consisting of: stress -> increased free sterol and TORC2 inhibition -> activation of LAM proteins (as demonstrated by Relents and co-workers previously) -> removal of free sterol -> return to unstressed state of PM and TORC2.
Comments
Fig 2A: Is detection of PIP/PIP2/PS linear for target, or possibly just showing availability that is increased due to local positive curvature?
Can any marker be identified for the D4H spots at 2 minutes? In particular, are they early endosomes (shown by brief pre-incubation with FM4-64)?
Is there any functional (& direct) link between Arp inhibition (as in the Pombe study of LAMs by the lab of Sophie Martin) and PM disturbance by amphipathic molecules ?
Minor
Fig 2A: Labels not clear. Say for each part what FP is used for pip2. Move fig s2d to main ms. The 1 min and 2 min data are integral to the story
The role of Lam2 and Lam4 in retrograde sterol transport has in vivo only been linked to one of their two StART domains not both, as mentioned in the text.
Throughout, images of tagged D4H should be labelled as such, not as "Ergosterol".
These results in budding yeast are likely to be directly applicable to a wide range of eukaryotic cells, if not all of them. I expect this paper to be a significant guid elf research in this area.
The paper specifically points out that the current experiments do not distinguish the precise causation among the two outcomes of stress: increased free sterol and TORC2 inhibition. Of these two outcomes which causes which is not yet known. If data were added that shed light on this causation that would make this work much more signifiant, but I can understand 100% that this extra step lies beyond - for a later study for which the current one forms the bedrock.
Note: This response was posted by the corresponding author to Review Commons. The content has not been altered except for formatting.
Learn more at Review Commons
We thank all the reviewers for their helpful and constructive comments and for their time.
Reviewer #1 (Evidence, reproducibility and clarity (Required)):*
Summary: Dady et al have developed fluorescent reporters to enable live imaging of cell behaviour and morphology in human pluripotent stem cell lines (PSCs). These reporters target 3 main features, the plasma membrane, nucleus and cytoskeleton. Reporter PSCs have been generated using a piggyBac transposon-mediated stable integration strategy, using a hyperactive piggyBac transposase (HyPBase). The same constructs were also used for mosaic labelling of cells within 2D cultures using lipofectamine transfection.
The reporters used are tagged with either eGFP or mKate2 (far red) and tag the plasma membrane (pm) via the addition of a 20 amino-acid sequence from rat GAP-43 to the N-terminus of the fluorescent protein, the nucleus via Histone 2B with a laser-mediated photo-conversion option (H2B-mEos3.2), and the cytoskeleton via F-Tractin. In total, the authors produced lines with the following:
• pm-mKate2 (far red) • pm-eGFP (green) • H2B-mEos3.2 (green to red) • F-tractin-mKate2 (far red) • H2B-mEos3.2 and pm-mKate2 (green to red, plus far red)
The cell lines used to generate these were the human embryonic stem cell line H9 and human induced pluripotent cell line ChiPS4. The constructs were also used to label cells in a mosaic fashion, using lipofectamine transfection of the original cell lines once they had formed neural rosettes.
Using these cells, Dady et al then performed live imaging in vitro of human spinal cord rosettes and assessed cell behaviour. In particular they analysed mitotic cleavage planes and apical positioning of neural progenitor cells (NPCs), and assessed actin dynamics within these cells. They showed a slowing of the cell cycle length after the initial expansion phase, an increase in the rate of asymmetric division of these NPCs, and abscission of the apical membrane during these divisions. The F-tractin reporter showed enrichment at the basal nuclear membrane during these cell divisions, suggested to help prevent basal chromosome displacement during mitosis.
Major comments: The data presented are convincing and could be strengthened by the following additions and clarifications:*
Manufacturer’s instructions for Lipofectamine 3000 transfection (6 well plate):
Cells in IBIDI dishes were left to develop in a sterile incubator overnight and mosaic fluorescence was observed the following morning (~16h post-lipofection).
Will these cell lines and constructs be made publicly available after publication?*
The cell lines can be made available: for those reporters made in the H9 WiCell line an MTA will first have to be signed between the requesting PI and WiCell and permission for us to share the line(s) confirmed by WiCell; similarly, for reporters in ChiPS4 line an MTA will first need to be signed between the requesting PI and Cellartis/TakaraBio Europe. We will need to make a charge to cover costs. Constructs will be deposited with Addgene.
In the Results we make clear that all lines created are polyclonal, with exception of a pm-eGFP ChiPS4 line, which is a monoclonal line (lines 145-150). We do not have direct data measuring cell proliferation but collected cell passaging data for all the reporter lines. This showed that they grow to similar densities at each passage compared to the parental line (this metadata is now provided as Supplementary data 1 and is cited in the Methods, line 348).
As a proof of principle for this approach, we created one monoclonal line from a polyclonal line ChIPS4-pm-eGFP. The latter was made by selecting an individual clone and this was then expanded and characterised for expression of pluripotency markers (immunocytochemistry data Figure S4), and the ability to differentiate into 3 germ layers (qPCR Supplementary data 1). This information is already cited in the Methods (Lines 358-362).
Minor comments: 1. Some images in the figures and supplemental movies are low in resolution, for example the DAPI in Fig 4B, making it hard to distinguish individual cells. Please increase this.
We consider the DAPI labelling in Figure 4b to be clear, however, we wonder whether the reviewer was expecting to also see this combined with the other markers. We have therefore now provided these merged additional images in a revised Figure 4.
This has now been provided in revised Figure 4B.
We have added indicative arrows to the movies, and note that more detailed labelling of the series of still images from these movies are provided in the main figures (Figures 3D and 4E & F).
*Reviewer #1 (Significance (Required)):
Human neurogenesis is currently poorly understood compared to many model systems used, yet key differences have already been identified between the human and the mouse, prompting the need for further investigation of human neural development. A major reason that human neurogenesis has been difficult to study is a lack of tools to enable cell morphology and behaviours to be analysed in real time.
The reporters and reporter PSC lines generated by Dady et al will allow many of these cell characteristics to be observed using live imaging. For example, the morphology of neural progenitors during and after cell divisions, how the apical and basal processes and membranes are divided, and how the actin cytoskeleton helps to regulate these processes.
*Importantly, PSC lines can be very heterogeneous, making generating reporter lines costly and time intensive. The use of these reporters with lipofectamine transfection, for a mosaic labelling, allows the visualisation of the plasma membrane, nucleus and cytoskeleton in any human PSC/NPC line, or even in human tissue cultures, without the need to generate each specific reporter line, making it a valuable tool for many labs in the field.
We strongly agree with this final point; this is a major reason for our study.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):*
The manuscript describes the generation of novel lines of human pluripotent stem cells bearing fluorescent reporters, engineered through piggyBac transposon-mediated integration. The cells are differentiated into neuronal organoids, allowing to capture cellular behaviors associated to cell division. A replating protocol allows the observation of aging neurons by reducing the thickness of the tissue thereby facilitating live imaging. The authors also leverage the transposon technology to create mosaically-labelled organoids which allows visualizing aspects of neuronal delamination, notably cytoskeleton dynamics. They discover an undescribed pattern of F-actin enrichment at the basal nuclear membrane prior to nuclear envelope breakdown.
L104-109: "Moreover, the transposon system obviates drawbacks of directly engineering endogenous proteins...". Despite the risk of endogenous protein dysfunction, directly tagging allows the full regulation of gene expression (including the promoter, the enhancers and other regulatory regions rather than a strong constitutive promoter such as CAG). In addition, the number of copies integrated and the genomic regions are variable with PB, which does not reflect the endogenous expression. This could be rephrased by nuancing the advantages and drawbacks of each approach. The PiggyBac method is easier and faster, but it results in overexpression of a tagged protein that will be expressed since the hESC state and might not reflect the expression dynamics of the endogenous protein.* We agree and have now revised this in the Introduction L109-118.
*L124-126: "To monitor cell shape and dynamics we used a plasma membrane (pm) localized protein tagged with eGFP or mKate2 (pm-eGFP or pm-mKate2)." Could the authors provide more details and a reference on the palmitoylated rat peptide use to force membrane expression? *
This information, including the peptide sequence, is provided in the Methods (L330-331), we have now added a reference addressing its role in membrane localisation PMID: 2918027.
L132-133: " Finally, to observe actin cytoskeletal dynamics we selected F-tractin, for its minimal impact on cytoskeletal homeostasis".
A recent JCB paper (https://doi.org/10.1083/jcb.202409192) suggests that "F-tractin alters actin organization and impairs cell migration when expressed at high levels". Whether the overexpression of F-tractin in hESC using a CAG promoter reflects the physiological F-actin dynamics and/or if the high levels could lead to an alteration of cell behavior should be addressed or at least discussed. The paper we cite in this sentence (Belin et al 2014) evaluates F-tractin expression against other approaches to labelling and monitoring the actin cytoskeleton and concludes that in comparison F-tractin has minimal impact.
We do appreciate that expression above the endogenous level has the potential to alter cell behaviour and have revised the paper to more explicitly acknowledge this: in the Introduction (L109-112), and in the Discussion/conclusion (L289-293) where we now note the recent advances reported in Shatskiy et al. 2025 PMID: 39928047.
“A further potential limitation of this approach is that over-expression driven by the CAG promoter might not reflect physiological protein dynamics and/or alter cell behaviour; for example, high levels of F-Tractin can impair cell migration and induce actin bundling, interestingly, this can now be minimised by removing the N-terminal region (Shatskiy et al 2025)”.
L146-147: "...to generate polyclonal cell lines selected for expression of easily detectable (medium level) fluorescence for live imaging studies". What are the criteria used to define medium level? Number of copies integrated into the genome? Or levels by FACS during clone selection?
To clarify, all the lines presented here are polyclonal, except for one clonal line, pm-eGFP in ChiPS4. The numbers of copies integrated may vary from cell to cell in polyclonal lines. In this study, we selected cells for all lines with a FACS gate and this data is presented in Figure S1 (see line 147).
L260-263: "Efficient stable integration and moderate expression levels were achieved by optimising, i) the quantity and ratio of piggyBac plasmids and transposase and ii) subsequent FACS to exclude high expressing cells, as well as iii) transfection methods, including temporally defined lipofection in hiPSC-derived tissues." The ration 5:1 is classically used for PB Transposase delivery, however there is still high variability in the number of copies integration. Lipofection in derived tissues has been shown to be challenging. Could the authors should provide quantitative data regarding the efficiency of their approaches, notably the level of mosaicism one could expect?
We provide quantitative data for the efficiency of transfection using nucleoporation assays (FACS data presented in Supplementary figure S1), which shows more than 80-90% efficiency for eGFP in 82.82% of cells, mKate2 in 92.74% of cells, and H2B-mEos3 22.75% of cells, while 13.79% of cells co-expressed pm-Kate and H2B-mEos3.2. No comparative data regarding the efficiency of the tissue Lipofection assay was collected: our goal was to label single/small numbers of cells in order to monitor individual cell behaviours, and this “inefficient labelling” was readily achieved following the manufacturer’s instructions (please see response to Review 1 point 1), further details are now provided in the Methods.
L191-194: "We further wished to monitor sub-cellular behaviour within the developing neuroepithelium. To achieve this, we devised a strategy to target a mosaic of cells in established neural rosettes using lipofection. PiggyBac constructs and HyPBase transposase were transfected into D8/D9 human spinal cord neural progenitors using lipofectamine (Felgner, et al., 1987)(Fig. 3A)." The mosaicism is not an all or nothing in this method but also leads to variations in expression levels among the positive cells. The protocol for lipofection could be better detailed to allow easy reproduction by other teams, and its expected efficiency should be discussed. It would be interesting to explore the relationship between individual cells phenotype and expression levels. Please see response to Reviewer 1 point 1 above for more detailed lipofection protocol which generated mosaic expression, this is now also included in the Methods. We agree that investigating the relationship between individual cell phenotypes and expression levels would be interesting, but we think this is beyond the scope of this paper.
Additional comments: -Did the authors perform karyotyping of the hPSCs prior to use in the differentiation protocol?
As these are polyclonal lines, we did not undertake karyotyping. This could be done for the one monoclonal line described here (pm-eGFP ChiPS4 line): we lack funds for commercial options, but we are exploring other possibilities.
-Were pluripotency assays performed after reporter lines generation?
These were carried out for the clonal pm-eGFP ChiPS4 line (lines 145-150). The latter was made by selecting an individual clone and this was then expanded and characterised for expression of pluripotency markers by IF (Figure S4), and the ability to differentiate into 3 germ layers by qPCR (Supplementary data 2). This information is provided in the Methods (Lines 358-362).
*-Did the authors measure the cell proliferation rate in H2B-overexpressing cells and controls? Since H2B plays an important role in cytokinesis, it could interfere in cell division when H2B is overexpressed (see doi: 10.3390/cells8111391). *
We did not directly measure cell division when H2B is over-expressed. However, we assessed cell -passaging time of all the transfected cell lines. This showed that they grow to similar densities at each passage compared to the parental line (this is now provided as Supplementary data 1 and is cited in the Methods, line 348). We also found no difference between apical visiting time of progenitors in spinal cord rosettes expressing pm-eGFP or H2B-mEoS3.2, further supporting the conclusion that levels of H2B-mEoS3.2 expression achieved in this line did not interfere with cell division (metadata provided in Supplementary data 3).
The authors should provide data concerning the efficiency of expression of the distinct markers after electroporation. This is provided in Supplementary Figure S1 (FACS data) and detailed above for this reviewer.
*At Fig 1C, the schematic representation describes clone selection, however in the methods it is stated (L348-349): "Sorted cells expressing medium levels of fluorescence were expanded and frozen then representative lots of each polyclonal cell line...". There is some confusion regarding which experiments were performed using polyclonal medium-level mixed populations or monoclonal populations. *
We apologise for any confusion and have revised the Figure 1C schematic to indicate that cells can be selected to either make polyclonal lines or clonal lines.
*Reviewer #2 (Significance (Required)):
The study provides novel tools, as well as elements regarding neuroepithelium biology. It is well conducted and written, and the quality of images is excellent. It reads more as a resource paper in its current version, since the observation regarding neural cell division and delamination are interesting but not deeply explored, so this review will focus on those technical aspects rather than the novelty of the biological findings.
This study would be of interests for researchers in stem cells and organoids, developmental biology, and neurosciences.
Reviewer #3 (Evidence, reproducibility and clarity (Required)):
In the manuscript, "Engineering fluorescent reporters in human pluripotent cells and strategies for live imaging human neurogenesis" the authors Dady et al. describe the adaptation of a recent advancement in transposase technology (HyPBase) as a method to integrate live reporters in human pluripotent stem cells. They show that these florescent reporters paired with new imaging strategies can be used to confirm the existence cellular behaviour described in other species such as the interkinetic nuclear migration (IKNM) of dividing progenitors in neural tube development. Finally, they demonstrate that this live imaging system is also able to discover novel biology by identifying previously undescribed actin polymerization at the basal nuclear surface of cortical progenitors undergoing cell division. Overall, the study presents two examples in which this adapted tool will aid in live-imaging studies of cellular biology.
Major Concerns: 1. This work needs more controls to properly demonstrate claims that their engineering strategy provides an advancement to current Piggyback methods. Their HyPBase strategy needs to be compared and quantified in terms of efficiency with other methods to support their claims (increased detection and reduced phototoxicity).*
We do not make specific claims for our experiments with respect to the superiority of HyPBase strategy. Our comments on this approach referred to by the reviewer here are in the Introduction (L 94-103), are supported by the literature (e.g. more stable gene expression than native piggyBac or the Tc1/mariner transposase Sleeping Beauty (Doherty, et al., 2012, Yusa, et al., 2011) and serve to explain our selection of HyPBase for our experiments. We make a case for using HyPBase as opposed to another transposase and although it would be interesting to compare efficiencies, this comment does not specify what “other methods” might be informative.
2.Throughout the manuscript more quantification is needed of the results. How many rosettes were examined? Were all the reported cells within one rosette? Were there differences between rosettes? This should be done for both the spinal and cortical differentiations.
The reviewer appears to have missed this information – we placed detailed quantifications in the figure legends (numbers of independent experiments and rosettes) and in the Methods in a specific section on Quantification of cell behaviour (L465-486), rather than in the main text. These has since been further updated and we now also provide additional metadata in the form of Excel spreadsheets for quantifications and analyses made for both spinal cord and cortical rosettes (Supplementary data 3 and 4 respectively).
Minor Comments: 1. Line 246 needs quantification shown in figures of the statements made. Specifically, how many cells were measured to get this number?
This information was provided in the figure 4 legend and we have since added numbers to these data; we were able to monitor 169 divisions in 21 rosettes; 154/166 divisions had vertical cleavage planes (symmetric) and 12/166 had horizontal cleavage planes (asymmetric).
These detailed observations were made in two independent experiments, along with observations of basal nuclear membrane F-Tractin localisation. This is noted in figure 4 legend, Methods and detailed metadata is provided in Supplementary data 4.
2.How many cells in the cortical rosettes had the enriched actin at the basal nuclear surface?
We confidently observed basal nuclear membrane F-Tractin enrichment in 141/146 divisions, for the remaining 20 cases (166-146), we could not tell whether F-Tractin is enriched or not at the basal nuclear membrane either because of low expression levels or because the basal nuclear membrane was out of focus at NEB. In 5 cases, we did not see the basal nuclear enrichment despite sufficient F-Tractin expression levels and the nucleus being in focus. We have updated the Fig4 legend excluding the non-analysable cases and see detailed metadata is provided in Supplementary data 4.
*Reviewer #3 (Significance (Required)):
General Assessment: This manuscript makes a very minor advancement in the field of stem cell engineering and developmental biology, but one that is worthy of publication with a few edits.
Advance: While PiggyBac reporters are widely used in stem cell engineering, Dady et al. demonstrate a new workflow using HyPBase which would be beneficial to the field. However, to increase this benefit, much more description and quantification of the methods would be needed. The biological advances of this manuscript are also very minor, but interesting as most of them confirm that human neural rosettes mimic many of the observed cell behaviours seen in animal models. Along these lines is the actin dynamics observation in cortical rosettes is interesting, but a preliminary observation and in need of follow up experiments.
Audience: Regardless, this technique would be of interest to the wider field of stem cell engineering.
My Expertise: Human Stem Cell Engineering, Neural Tube Development*