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
_Below we address all the comments by the reviewers. However, the figures that were used in our response are unfortunately not displayed in this format. _
Reviewer #1
Evidence, reproducibility and clarity
Thanks to the development of Ribo-Seq, translational buffering has been reported in the database of published Ribo-Seq and matched RNA-Seq, Rao et al. attempt to understand the mechanism underlying translational buffering of mRNA variation across diverse materials. Although the authors' report provides a step forward in our understanding of translational buffering, this reviewer found a series of concerns in this paper. These points could be tackled to improve the reliability of their findings, the strength of their main message, and the global understandability of the paper.
Major comments: 1. This paper heavily relies on the reference 18. However, this paper was not properly stated (no page or journal number); the study in Bioinformatics is nowhere to be found on the website, despite being out in 2024 apparently. Either title is wrong (yet a biorxiv can be found). This reviewer guessed that the reference 18 may be accepted. However, without a proper reference, this paper could not be judged since nearly all the parts of this work have been based on the reference 18. Also, the Ribobase data used in this manuscript comes from this reference, so it had better be well defined, especially when another Ribobase data set seems to be available online: http://www.bioinf.uni-freiburg.de/~ribobase/index.html
We apologize for the citation issue. This citation by Liu et al , 2024 (18) was a preprint from BioRxiv. This manuscript is now published in Nature Biotechnology. The reference has been updated in the revised version of the manuscript. The reference number in revised manuscript is Liu et al, 2025 (23).
In the Discussion, the authors mentioned "TE is based on a compositional regression model (18) rather than the commonly applied approach of using a logarithmic ratio of ribosome occupancy to mRNA abundance." This important information should be mentioned in the early section of the manuscript. Related to this, there are other published methods for exploring change in translation efficiency (e.g., 10.1093/bioinformatics/btw585; 10.1093/nar/gkz223) that could also be suitable in this context. It is not entirely clear if their approach is better than before. Again, the improper reference to 18 made our assessment of this work difficult.
We apologize and acknowledge the impact of the citation issue on this point. In Liu et al (2025), we have provided a comparison between our approach and the log-ratio strategy. We also agree that additional context was needed within the current study. Hence, we have now included more detailed information about the TE calculations in the initial results section (line 94).
As noted by the reviewer, several other methods have been developed previously for measuring changes in translation efficiency. These methods are designed to be used in cases of paired designs where there is a treatment or manipulation that is assayed along with controls. While these methods are highly valuable in assessing differential TE, they are unable to accommodate the type of meta-analyses described in our study. In particular, we do not report changes/differential TE with respect to a control sample but instead focus on the coordinated patterns of TE across experiments. We now note this important distinction in the manuscript in the discussion section (line 494).
The paper mainly relies on detecting a set of buffered genes using mRNA-TE correlation and MAD ratios (Ribo-Seq/RNA-Seq). While the concept seems sound, the authors should ensure that this method is reliable. Several controls could be used to confirm this. First, if any studies in humans or mice have described a set of genes as buffered, it would be worth checking for overlap between the authors' set of 'TB high' genes and the previously established list. Furthermore, the authors could use packages explicitly developed for translational buffering detection, such as annota2seq (https://academic.oup.com/nar/article/47/12/e70/5423604?login=true). Not all of the data used by the authors may be suitable for such packages, but the authors could at least partially use them on some of their datasets and see whether the buffered genes reported by these packages match their predictions.
We thank the reviewer for this constructive suggestion. To the best of our knowledge, no prior study in humans or mice has systematically analyzed translational buffering across a wide range of conditions. As a result, defining a gold-standard set for benchmarking is currently not feasible.
While packages such as anota2seq have proven highly valuable for identifying buffering effects in controlled experimental designs (e.g., comparing a treatment to a matched control), they are not readily applicable to the type of large-scale meta-analysis we present here.Our study integrates ribosome profiling and RNA-seq data across diverse datasets and conditions, which lies outside the design scope of such tools.
The most relevant point of comparison to our work is Wang et al. 2020 Nature, which examined a related but distinct form of translational buffering across species for a given tissue. We now present the overlap of genes identified as buffered in our study vs Wang et al. 2020. The details are presented in the reviewer's comment 5-2.
The threshold of 'TB high' or 'TB low' (top and bottom 250) is somewhat arbitrary. Why not top 100 or 500? The authors should provide a rationale for this choice. Also, they could include a numeric measure of buffering (the sum of the two rankings is probably suitable for this purpose). Several of the authors' explorations are suitable for numerical quantification (GO enrichment can be turned into GSEA, and the boxplot can be shown as correlations)
Thanks for these suggestions. We agree that the threshold used to define TB high and low are somewhat subjective. We ensure that changing this cutoff as suggested is easily achievable with the provided R script. These can be used to reproduce all of the reported analyses of translational buffering with different cutoffs.
To further assess whether our conclusions are robust to the selection of these thresholds, we tested several different values to define the TB high and TB low groups. As an example, we show here that the effect on protein variation and association of intrinsic features like the UTR lengths with the buffering potential of genes for different thresholds (i.e. if the TB high = top 100 or TB high = top 200) remain similar to the current cutoff of 250. However, if we increase the cutoff of TB high to 2000 and TB low to top 2000-4000 , the difference between the various features is diminished (Figure A& B). Further, protein variation (human cancer cell line and tissue) also becomes more similar across the three categories, possibly indicating a reduced regulatory potential of genes as their rank increases (Figure C& D).Our analyses reveal that highly ranked genes show associations with particular features, indicating an underlying hierarchy in translational buffering potential. This point is now discussed in the manuscript (line 177).
Legend: Effect of different thresholds on . A. Length features B. Median RNA expression C. Protein variation in human cancer cell line and D. on Primary human tissues
In response to the reviewer's suggestion of presenting data using numerical quantitation, we incorporated several additional inclusions in the manuscript.
- We now report association of CDS / UTR length with translational buffering as a function of their translational buffering rank with highly ranked genes showing associations with particular features, indicating an underlying hierarchy in translational buffering potential (Sup Fig 3 A-B) Ii. We now include scatter plots which show that highly ranked genes have lower variation at the protein level in both cancer cell line and primary tissues (Sup Fig. 6 A-C).
Iii. We have now carried out modified GO enrichment analyses. Specifically, Gene Ontology enrichment analysis was performed for the TB high genes in humans and mouse using the clusterProfiler R package. Lists of TB high genes in human or mouse were analyzed against the Gene Ontology (GO) database using the enrichGO() function, with the organism-specific annotation database (org.Hs.eg.db for human or org.Mm.eg.db for mouse) as reference. Gene identifiers were supplied as gene symbols, and all genes in the current study were used as the background universe. Enrichment was carried out for the Biological Process (BP) ontology, with significance assessed by the hypergeometric test. P-values were adjusted for multiple testing using the Benjamini–Hochberg method, and terms with an adjusted p-value Legend: Gene Ontology (GO) enrichment analysis of the TB high gene set, performed with the clusterProfiler R package. Enriched GO Biological Process terms are shown after redundancy reduction using clusterProfiler::simplify. Each dot represents a GO term, with dot size indicating the number of genes associated with the term and color reflecting the adjusted p-value (Benjamini–Hochberg correction). Only the top non-redundant terms are displayed.
- *
Additionally, we performed Gene set enrichment analysis using the list of genes ordered according to their RNA-TE correlation. Hence lower ranks have lower RNA-TE correlations. The GSEA plots show significantly enriched Gene Ontology Biological Process (GO:BP) terms at the lower ranks of the ordered gene list. Together, these analyses further emphasize the observation that genes involved in macromolecular complexes are translationally buffered.
- *
Legend: Curves represent the enrichment score (ES) across the ranked gene list, with vertical bars indicating the positions of pathway-associated genes. The enrichment was identified using the gseGO() function from clusterProfiler.
Several of the statements of the authors in the Introduction or Discussion sections are not entirely true regarding the literature on the topics, or lack major papers on the topic, and therefore, they are a bit misleading. Among others, here are some:
We thank the reviewer for the suggestions and now have been incorporated in the revised manuscript, accordingly.
5-1 "In addition, genetic differences arising from aneuploidy, cell type differences or variability observed in the natural population can further determine the amplitude of variation (4-7). The effect of mRNA variation under these conditions is mostly reflected at the protein levels (2, 4-8).". Several recent or more ancient papers suggest that mRNA variation coming from aneuploidy, natural genetic variation, or CNV is buffered or not well reflected at the protein level: DOI: 10.1038/s41586-024-07442-9 DOI: 10.1073/pnas.2319211121 DOI: 10.1016/j.cels.2017.08.013 DOI: 10.15252/msb.20177548
We agree that mRNA variation coming from aneuploidy, natural genetic variation, or CNV is buffered or not well reflected at the protein level for some genes. This point has now been revised in the introduction. We have incorporated all the suggested literature into the revised manuscript (line 38).
5-2: The authors should also consider mentioning these studies and softening their initial statement. "Similarly, translational buffering of certain genes have been reported in mammalian cells, specifically under estrogen receptor alpha (ERα) depletion conditions (16).". Translational buffering has been deeply explored in mammalian tissues and even across several mammalian species in this study (DOI: 10.1038/s41586-020-2899-z). In this, the authors also provide a nice exploration of the gene characteristics that are associated with translational buffering. The authors should mention it and compare the study's findings to theirs ultimately.
We thank the reviewer for this suggestion. We have now cited the recommended study in the revised manuscript (line 65). Here, we provide a comparison of its findings with ours. While this related work offers important insights into translational buffering, its focus is on buffering across species within a given tissue, whereas our study emphasizes buffering across conditions, cell types, and treatments within a species. Despite this difference in focus, the comparison is highly informative, and we now highlight both the similarities and distinctions between the two studies in the relevant section of the revised manuscript.
Wang et al. calculate the variation at the transcriptome level vs at the translatome level and is represented as delta ∆ value for each gene. A lower value represents lower variation at the ribosome occupancy level than at the mRNA levels across various species. We classified the genes in the Wang et al study as TB high, TB low genes or others as identified in the current study while indicating the calculated delta ∆ from Wang et al. Many of the genes with a lower delta value (are delta ∆ Legend: A. Dot plot to highlight the delta value of all genes in the Wang et al study (also present in RiboBase) which are further grouped as TB high, low or others in (A) brain and (B) liver.
5-3: "Differences in species evaluated and statistical methods have resulted in conflicting interpretations (13, 28).". These conflicting results have been previously discussed in reviews on the topic that would be worth mentioning: DOI: 10.1016/j.cell.2016.03.014 DOI: 10.1038/s41576-020-0258-4
We have added these reviews at the appropriate location of the manuscript.
- In addition to the p-values stated in the main text, the authors should annotate their plots when they find significant differences between groups to greatly facilitate the visual interpretation of the graphs.
We have now annotated many of the relevant graphs with p-values to facilitate visual interpretation, adding them where space and figure design allow.
Based on the data of Figure 4D, apparently, ribosome occupancy was not buffered even in high TB sets. The authors may argue that translational buffering may not cope with such a strong mRNA reduction. In that case, how big a difference in mRNA level does the buffering system adjust in protein synthesis? The authors should test gradual gene knockdown and/or overexpression and conduct Ribo-Seq/RNA-Seq to survey the buffering range.
We appreciate the reviewer’s suggestion regarding the experiment to determine the buffering range.To understand this for multiple genes, we attempted a series of knockdowns using CRISPR/gRNA approach using a MutiCas12a approach. We targeted 8 buffered and 2 non-buffered genes using a 10-plex crRNA along with 10-plex gRNA serving as a negative control (Figure below). The fold change at the mRNA level of the targeted gene was within the variation range observed in replicates for other non-targeted genes. The challenge in performing a gradual knockdown is the subtle changes in RNA expression falls within the margin of error of estimation, making it difficult to understand the clear implications of the mRNA levels on buffering. Hence, the precise experimental manipulation of mRNA expression levels that would be conducive to translational buffering remains highly technically challenging. As noted in our manuscript (Figure 4D), the conventional approaches for manipulation of transcript abundance lead to larger changes than typically observed as a result of natural variation.
*Legend: Validation of translational buffering by targeted knockdown of genes. A. The scatter plot shows the coefficient of variation of mRNA and ribosome occupancy between HEK293T cells targeted with sgRNA of different efficiencies. The genes indicated in blue are buffered and those in green are non buffered genes. B. The plot shows the fold change in mRNA abundance and ribosome occupancy as compared to cells that were infected with non-targeting crRNA array control (ratio of cpm in test vs control). Each color represents a gene and each point of a gene represents cells targeted by one of the four CRISPR arrays. *
"differential transcript accessibility model" could not be functional if mRNA is reduced beyond the accessible pool (i.e., less than the threshold, all the mRNAs are translated without buffering). The authors should carefully reconsider this model and the effective range of mRNAs.
We agree with the reviewer that according to the 'differential transcript accessibility model,' transcripts with abundances below a certain threshold should be completely accessible to the translational pool. Further, this could also be true for the other model, wherein initiation rate cannot increase beyond a particular threshold for transcripts of very low abundance. However, our observation from our haploinsufficiency analysis (Figure 4 B& C) and siRNA knockdown analysis from RiboBase (Figure 4 D) suggests that buffering might be possible within a given range of transcript abundance. Testing the buffering range by serial knockdowns might help in determining the threshold at which transcripts exhibit buffering. However, due to the challenges of serial knockdown as discussed above, makes this analysis difficult with Ribosome profiling and matched RNA-seq approach. An alternative approach could involve imaging translating and non-translating mRNA of buffered genes in different cells, which may help distinguish the two models. However, this falls outside the scope of the manuscript.
Minor comments:
- Some figures are of poor quality as they seem to have points outside of the panel representations... Like Figure 3C, one point is out of the square, same for Figure 4E. Similarly, on figure 5F, some outliers seem to be clearly cut from the figure (maybe not, but then the author should put a larger space between the end of the figure and the max y points). Same for panel S2D and S6D, this does not sound so rigorous.
We agree and apologize for this issue. The axes of the figures have been annotated appropriately to indicate the presence of outliers in the figures.
- There are several typos or weird sentences. Here are some (but maybe not all): 2-1: [...]with lower sums corresponding to higher final ranks. "two rankings". Based on these final ranks[...] 2-2: For each dataset, median absolute deviation (MAD) "i" protein abundance was calculated across samples 2-3: [...]neighbor method implemented in the MatchIT package (38) Differences in protein[...] a point is missing here. 2-4: Additionally a second dataset providing predictions of haploinsufficiency (pHaplo score) and triplosensitivity (pTriplo score) for all autosomal genes (25) was used to asses the distribution of these score"S" across buffered and non-buffered gene sets . There is a missing "s" at "score" and there is a space between the last word and the final point.
The necessary corrections have been incorporated in the revised version of the manuscript.
- In the "Lymphoblastoid cell line data analysis:" section, this reviewer wonders why the authors used a different method to calculate buffering compared to before.
The main reason is the limited sample of the lymphoblastoid cell line data. In our larger analyses, we could use median absolute deviation as a robust metric of dispersion across heterogeneous samples. However, given the smaller dataset in that study we decided CV would be a better indicator of dispersion. To evaluate the potential for translational buffering of genes from RiboBase, we used two metrics. The first was the negative correlation between translation efficiency and RNA abundance across samples. The second metric relied on the ratio of variation in ribosome occupancy to variation in RNA levels. Given the limited sample size of the lymphoblastoid cell line dataset, we used the coefficient of variation (CV) instead of the median absolute deviation (MAD), as the data in this study were normalized using counts per million (CPM) rather than the centered log-ratio (clr) normalization used in RiboBase. This CV ratio allowed us to assess the effect of natural variation in RNA abundance on ribosome occupancy.
- "Samples which had R2 less than 0.2 were removed as the residuals calculated for these samples could be unreliable". These samples for which the correspondence between RNA-Seq and Ribo-Seq is low wouldn't be the ones most impacted by translational buffering? Is it sure that the authors are not missing something here?
We agree with the reviewer that genes that show translational buffering may not conform to linear relationships between the two parameters. However, the proportion of genes exhibiting this buffering effect is not expected to significantly influence the overall regression fit. Instead, we hypothesized that low quality samples or truly different relationships between the two parameters can make this relationship nonlinear, rendering it unsuitable for linear regression analysis for calculation of TE.
To address these possibilities, we first analysed a commonly used proxy for data quality. Given the characteristic movement of ribosomes across mRNAs, periodicity of sequencing reads is a useful metric to assess whether reads are randomly fragmented, as in RNA-seq, or specifically represent ribosome-protected footprints. For this, we compared two groups: samples that were removed (~30) and those retained for analysis. We plotted the distribution of periodicity scores for all samples in both groups. For the calculation of periodicity scores, first the percentage of reads mapped to the dominant frame position across the dynamic ribosome footprint read length range was calculated for each sample. The periodicity score was calculated by taking the weighted sum of these dominant percentages, with weights based on the total read counts at each length.
The results indicate that the removed samples did not have lower periodicity scores, suggesting that their quality in terms of periodicity was comparable to the retained samples.
To assess the second possibility, we checked if the study involved major perturbations, which may skew the relationship towards non linearity. The 30 samples that were removed came from 14 unique studies, 18 of which involved perturbation which possibly affected either of the two parameters. In addition to the genetic/pharmacological perturbations specific to the study, the overall conditions of the cells during an experiment could influence this relationship. Another point to note is that many of the filtered-out samples are HeLa and HEK293T cells, which show a normal relationship between ribosome occupancy and RNA abundance for the majority of cases.
These considerations suggest that removing these samples is most appropriate, as their inclusion could bias the TE calculations.
For Figure 4B and 4C, the authors should provide statistical tests and p-values to confirm the observed trends.
The haploinsufficiency and triplosensitivity analyses are now supported by a chi-squared test. The details of the statistical test are now mentioned in the text and the p-values have been noted on the respective figures.
In Figure 2A, the "all genes" color doesn't correspond to the point color.
The color in the figure has been modified in the revised version of the manuscript.
- "To understand if codon usage patterns are[...]". This comes slightly out of the blue. The authors could maybe explain why codon usage should be explored for translational buffering. The authors should cite recent key works in the fields: DOI: 10.1016/j.celrep.2023.113413 DOI: 10.1101/2023.11.27.568910
We would like to thank the reviewer for their suggestion. The references have been incorporated in the revised version of the manuscript. We have now explained why codon usage could be a contributor in determining the translational buffering potential (line 190).
"The change in each metric was calculated by subtracting the mean value in the control samples from that in the knockdown samples. This yielded the differential mRNA abundance and ribosome occupancy resulting from gene knockdown.". This looks statistically weak. The authors should consider using more robust methods like DESeq.
We thank the reviewer for the suggestion. We reanalyzed the selected studies using edgeR and the modified figure is included in the revised version of the manuscript (Figure 4D). The conclusion after this analysis remains essentially the same. In particular, translational buffering is ineffective when mRNA abundance is perturbed drastically. Additionally, the limited number of experiments with direct perturbation of buffered genes limit the generalizability of this observation. This limitation is included in the result section (line 342).
Legend: Scatter plot represents log2 fold change in RNA abundance and ribosome occupancy. Each point represents a gene and the fold change in its RNA and ribosome occupancy with respect to their controls. The line represents the line of equivalence. Buffered genes do not show less change in ribosome occupancy upon reduction in their RNA levels than other genes.
- "Genes in the buffered gene set had a higher codon adaptation index than the non-buffered set, indicating that candidates in the buffered gene set are relatively well expressed due to the presence of a higher proportion of the codons observed in highly expressed genes". What do the authors mean by "relatively well expressed"? Abundantly expressed? This sentence and the causality under it is unclear and should be modified or better explained.
We thank the reviewer for pointing out the lack of clarity in the sentence. We have now quantitatively measured the CAI in the three categories and modified the sentence to better explain the rationale in the revised version (line 183). “To understand if codon usage patterns are associated with translational buffering, we next analyzed codon properties across buffered and non-buffered human gene sets. The codon adaptation index quantifies how closely a gene’s codon usage aligns with that of highly expressed genes. Genes in the buffered gene set had a higher codon adaptation index than the non-buffered set. Specifically, 28.4% of TB high genes, 14% of TB low genes and 9.3% of genes in the other category fall within the top decile (>90th percentile) of codon adaptation index.”
The panel 4D is unclear. Is one point associated with one gene? Or is it the average of several genes? If it's one point for one gene, it is important to clearly state it because the number of cases is therefore quite low, especially for the TB high and low.
Each point and line are associated with a single gene. This is now clarified in the legend of the figure (line 364). The number of genes in this analysis is limited to the available ribosome profiling data with gene knockdown experiments.
- In Figure 2J, GGU (Gly), AAG (Lys), and ACU (Arg) provide negative effects on prediction, although these were enriched in the high TB set (Figure 2E). This contradiction should be explained.
While this appears to be a seeming contradiction, it is in line with what we expected. In particular, the objective of Figure 2J is to illustrate the features that predict the mRNA–TE correlation of genes, as identified using a LGBM model. The Spearman correlation shown reflects the relationship between each feature and the mRNA–TE correlation values. A negative correlation for codons such as GGU (Gly), AAG (Lys), and ACU (Thr) suggests that enrichment of these codons is associated with lower mRNA–TE correlation. This is in agreement with our observation in Figure 2E which suggests that high TB genes are enriched in these codons. In contrast, transcript size exhibits a positive correlation, indicating that shorter transcripts tend to have lower mRNA–TE correlation values.
Given that the choice of colors is a potential source of confusion, we have revised the text (line 230) and the figure (& legend) to try to clarify this relationship.
The subtitle of "Translationally buffered genes exhibit variable association kinetics with the translational machinery in response to mRNA variation" sounds unfair to this reviewer. Since the authors did not work on kinetics directly, the use of this word is misleading.
We agree and revised the subtitle to “The association of translationally buffered genes with the translational machinery varies in response to changes in mRNA abundance"
- The explanation of Figure 5A "We next explored the potential mechanisms that may give rise to translational buffering. Specifically, we considered two non-mutually exclusive models by which mRNA abundance might be decoupled from ribosome occupancy. In the first, the "differential transcript accessibility model", mRNA abundance determines the fraction of transcripts that are accessible to the translational pool. In this scenario, an increase in mRNA abundance would be accompanied by a proportionally smaller increase in the fraction of transcripts entering the translating pool for buffered genes, compared to non-buffered genes. In the second, the "initiation rate model", the rate of translation initiation per transcript scales inversely with mRNA abundance. Under this model, the proportion of mRNA entering the translational pool would be comparable across buffered and non-buffered genes (Fig 5A)." is hard to understand. The authors should rewrite for a better understanding of the readers.
This section has been rewritten in the revised version of the manuscript. The text now reads as
“We next explored the potential mechanisms that may give rise to translational buffering. Specifically, we considered two non-mutually exclusive models by which mRNA abundance might be decoupled from ribosome occupancy. In the first, the “differential transcript accessibility model”, mRNA abundance determines the fraction of transcripts that are accessible to the translational pool. In this scenario, an increase in mRNA abundance would be accompanied by a proportionally smaller increase in the fraction of transcripts entering the translating pool for buffered genes, compared to non-buffered genes. In the second, the “initiation rate model”, the rate of translation initiation per transcript scales inversely with mRNA abundance. Under this model, as mRNA abundance increases, translation initiation on each transcript is reduced, thereby lowering the number of ribosomes per transcript. However, this mechanism allows a proportional increase in transcripts entering the translational pool for buffered genes, similar to non-buffered genes”
Significance
Thanks to the development of Ribo-Seq, translational buffering has been reported in various works. However, the systematic investigation has remained challenging. Employing the database of published Ribo-Seq and matched RNA-Seq, Rao et al. attempt to understand the mechanism underlying translational buffering of mRNA variation across diverse materials. A group of mRNAs whose expression variance is buffered at the translation level was comprehensively surveyed in humans and mice. The authors found a series of features in the translationally buffered genes, including high GC contents in the 5′ UTR, optimal codon usage, and mRNA length. The depletion or increase of one allele of the genes in the group may be particularly detrimental to cells. The authors' report provides a step forward in our understanding of translational buffering, appealing to the broad scientific community in basic and applied biology. However, this reviewer found a series of concerns in this paper, including clarity in the methods, experimental validation, referring the earlier works, etc. These points could be tackled to improve the reliability of their findings, the strength of their main message, and the global understandability of the paper.
We thank the reviewer for noting the significance of the work and for their constructive feedback.
Reviewer #2 (Evidence, reproducibility and clarity (Required)):
In this manuscript, Rao and colleagues present a comprehensive analysis of translational buffering in human and mouse by mining 1515 matched ribosome profiling and RNAseq datasets from diverse tissues and cell lines. They define translational buffering as genes whose TE is negatively correlated with mRNA abundance across conditions, and further identify candidates by comparing median absolute deviations of ribosome occupancy versus mRNA levels. The authors find a conserved set of buffered genes enriched for components of multiprotein complexes, demonstrate that buffered genes exhibit lower protein variability and greater dosage sensitivity, and propose two non-mutually exclusive mechanistic models (differential accessibility and initiation rate modulation). Finally, they perform complementary fractionation experiments in HEK293T cells to support these models.
These findings propose a novel, conserved mechanism of translational buffering that tunes gene expression in mouse and human, showing how intrinsic sequence features and cellular context cooperate to stabilize protein output across diverse conditions. However, further evidence is required to fully support the authors conclusions, particularly direct validation of the proposed models of buffering.
We thank the reviewer for their positive assessment and thoughtful suggestions that we address below.
Below are my main concerns:
-
The choice of the top 250 genes by spearman correlation and MAD ratio as "TB high" seems arbitrary. The authors should justify these cut offs (via permutation analysis or FDR control) and show that conclusions are robust to different thresholds.
We agree that the threshold used to define TB high and TB low is somewhat subjective, and we now clearly acknowledge this in the discussion section (line 485). We now provide an R script that reproduces all analyses of translational buffering, where changing this cutoff to higher or lower values is straightforward.
To ensure the robustness of our conclusions, we evaluated several thresholds for defining TB high and TB low. We observed that the conclusions hold within a reasonable range of values (100-250). For example, the effects on protein variation and the association of intrinsic features such as UTR lengths with buffering potential remain consistent when TB high is defined as the top 100 or the top 200 genes, compared with the current cutoff of 250. In contrast, when we define TB high as the top 2000 and TB low as ranks 2000–4000, the difference between the various features is diminished (Figure A& B). Further, protein variation (human cancer cell line and tissue) also becomes more similar across the three categories, possibly indicating a reduced regulatory potential of genes as their rank increases (Figure C& D). Our results show that highly ranked genes consistently associate with specific features, suggesting an underlying hierarchy in translational buffering potential.
Legend: Effect of different thresholds on . A. Length features B. Median RNA expression C. Protein variation in human cancer cell line and D. on Primary human tissues
The modified compositional regression approach for TE and imputation of missing values are central to the study, but details are relegated to supplemental methods. The manuscript would benefit from a clear comparison of this method against standard log-ratio TE estimates, including sensitivity analyses to missing-data imputation strategies
We thank the reviewer for the feedback. We have now added further description of the modified compositional regression and the imputation strategy in the results section (line 94). Comparison to standard log-ratio TE estimates and their limitations has already been detailed in Liu et al. 2025, Nature Biotechnology. Therefore, in the current manuscript we specifically focus on the effect of the imputation strategy.
Specifically, the modified imputation slightly improved concordance between the set of genes that are identified to be translationally buffered using the negative RNA-TE relationship or using RNA -Ribosome occupancy correlation (0.91 to 0.94). Further, we assessed the correlation between TE and protein abundance as measured by mass spectrometry from seven human cell lines (A549, HEK293, HeLa, HepG2, K562, MCF7 and U2OS). The protein measurements were obtained from PaxDb. The new imputation strategy slightly increased mean correlation between the TE and proteome abundance as compared to naive strategy. It specifically showed improved correlation for HepG2, A549 and HeLa cell lines. 3507 genes were used for this analysis that were common between PaxDb, Liu et al., 2005 and the current study.
Legend: Proteomics vs TE correlation of cell types without or with imputation strategy. Spearman correlation between compositional TE calculated as calculated by Liu et al., 2025 from 68 samples from 11 studies (HEK293), 86 samples from 10 studies (HeLa), 58 samples from four studies (U2OS), 29 samples from five studies (A549), five samples from two studies (MCF7), seven samples from two studies (K562) and 10 samples from two studies (HepG2) or from the current study. 57 samples from 10 studies (HEK293), 82 samples from 9 studies (HeLa), 58 samples from four studies (U2OS), 29 samples from five studies (A549), 5 samples from two studies (MCF7), one samples from one studies (K562) and 9 samples from two studies (HepG2) . 3507 genes were used for this analysis that were common between Paxdb, Liu et al., 2005 and the current study.
Human data are derived mainly from immortalized cell lines, whereas mouse data are from primary tissues. Pooling these heterogeneous sources may conflate cell type-specific regulation with intrinsic buffering. The authors should either stratify analyses by context or demonstrate buffering signatures remain consistent within more homogeneous subsets
We thank the reviewer for the suggestion and agree that heterogeneity could potentially mask cell type-specific buffering effects. The TB-high genes we report are those that show consistent and robust expression across diverse contexts. However, unlike RNA-seq datasets, the current number of ribosome profiling samples per cell type is still limited, and a more comprehensive assessment of context-specific buffering will require larger datasets that will accumulate over time.
Nonetheless, we have stratified the analysis by cellular context. Specifically, we grouped samples of the same cell-type and repeated the buffering analysis. We provide a new table listing TB-ranks of genes for the five cell types with the largest sample sizes as a table in github.
As an additional control, we compared buffering patterns between related and unrelated cell lines. For example, the correlation of TB ranks between related cell lines HEK293T (n = 98) and HEK293 (n = 57) is higher (0.46) than between either and an unrelated cell line, HeLa (n = 82). Similarly, the correlation between two liver cell lines, Huh7 (n = 39) and HepG2 (n = 9), is higher (0.20) than between Huh7 and a similarly sampled but unrelated lymphoblastoid cell line (LCL, n = 9; correlation = 0.05). While these analyses suggest that cell type-specific patterns may exist, their exploration is currently limited by sample size, as detecting buffering requires substantial variability in mRNA expression. We now highlight this as a limitation in the Discussion section (line 573).
*Legend: Spearman correlation between TB ranks of different pairs of cell lines. The first set indicates comparison with HEK293T. The second set indicates comparison between liver cells (HepG2 and Huh 7). *
The HEK293T fractionation experiments offer preliminary support for both the "accessibility" and "initiation" models, but only slope analyses are shown. To validate these models, the authors should perform targeted reporter assays (dual luciferase constructs with 5′UTR swaps) or manipulations of initiation factors (eIF4E knockdown) to directly test how transcript abundance alters initiation rates versus pool entry
We thank the reviewer for suggesting experiments to validate the proposed models. In the luciferase reporter experiments, constructs bearing the endogenous UTRs from non-buffered genes would be expected to result in expression that is proportional to transcript abundance. In contrast, swapping a 5’ UTR from buffered genes would mitigate this effect of translation buffering via “initiation rate model” depending on the 5 UTR sequence of transcript. However, as outlined below, this experiment has important caveats:
- Role of coding sequence: Such assays primarily test the contribution of the 5′UTR and do not address potential cooperative effects between the 5′UTR and the coding sequence (CDS). Thus, if 5′UTRs fails to recapitulate translational buffering, it would be unclear whether the buffering requires coordinated action of the 5′UTR and CDS or whether the gene in question simply does not conform to the initiation-rate model.
- Sensitivity of measurements: Reporter-based measurements often rely on RT-qPCR to quantify expression changes. While suitable for large fold-changes, small shifts may fall within the assay’s technical margin of error, limiting the interpretability of the results. iii. Gene-to-gene variability: Buffered and non-buffered transcripts likely span a wide range of intrinsic initiation rates. Selecting only a few “representative” transcripts for 5′UTR swapping could yield results that are not broadly generalizable.
Similarly, knockdown of general initiation factors will likely impact on both buffered and non-buffered genes, which could limit the ability to distinguish the effect of transcript abundance on translational buffering via either of the proposed models. We envision an alternative future approach that would involve single molecule imaging translating and non-translating mRNAs of buffered and non-buffered genes under varying abundance conditions in a physiological context. Such experiments are likely the most suitable for disentangling the contributions of accessibility versus initiation. While we find this an exciting direction for future work, it lies beyond the scope of the present manuscript.
The conclusion that buffering reduces protein variability relies on mass-spec comparisons, but ribosome occupancy does not always reflect functional protein output (due to elongation stalling or co-translational degradation). Incorporating orthogonal measures, such as pulse-labeling or western blots for key buffered versus non-buffered genes, would strengthen the link between buffering and proteome stability
We agree with the reviewer’s concern and have been acknowledged as a limitation in the discussion section. To address this with orthogonal approaches, we carried out several additional experiments. Specifically, we identified a study from RiboBase (GSE132703) that exhibited significant variation in FUS transcript (a translationally buffered gene) abundance across conditions—namely HEK293T wild type, LARP1A single knockout (SKO), and LARP1A/B double knockout (DKO) using their RNA-seq data. We reached out to the authors of the study and obtained these knockout cell lines. We reanalyzed RNA abundance under the different conditions by RT-qPCR and assessed protein levels by Western blot. Despite observing differences in RNA abundance, FUS protein levels did not exhibit corresponding change at the protein level.
We also selected a non-buffered gene; DNAJC6, that also showed RNA-level differences. However, the change in RNA expression was not consistent at the protein level. Some caveats of Western blot is its limited sensitivity which may prevent detection of subtle changes and that the measurements are steady-state protein levels which cannot resolve whether differences arise from altered synthesis or degradation.
*Legend : Validation of buffering gene by western blot: A. Plot showing the RNA abundance and ribosome occupancy of buffered gene ; FUS and non buffered genes; DNAJC6 with variation in HEK293T-wild type, LARP1A single knockout and LARP1A/B double knockout. B. Validation of the RNA seq data by qPCR. C. Western Blot showing the FUS, DNAJC6 and Actin in wild type and different mutants. D. Bar plot showing the quantification of western blot. *
In addition to this targeted analysis , we performed quantitative mass spectrometry to evaluate the effect of mRNA variation at the protein level at global scale.
LC MS/MS analysis was performed on the above samples in triplicates at the Proteomics facility of the University of Texas. A total of 4,048 proteins were identified using a peptide confidence threshold of 95% and a protein confidence threshold of 99%, with a minimum of two peptides required for identification. Total precursor intensities for all peptides of a protein was summed and was used for protein quantification using DEP (Differential Enrichment of Proteomics Analysis) Package, in Bioconductor, R (https://rdrr.io/bioc/DEP/man/DEP.html). DEP was used for variance normalization and statistical testing of differentially expressed proteins. As expected LARP1 protein was identified in the control cells but not in the single or double knockouts.
We then plotted the fold change in RNA as determined by edgeR analysis of RNA-seq from (Philippe et al. 2020) and the fold change in protein abundance from our mass spectrometry data. We observed that genes in the TB high group show reduced changes at the protein level compared to TB low or others as determined by the linear regression analysis in both single and double LARP1 KO mutants. This finding is consistent with our findings that buffered genes show lower variation in the protein abundance in response to change in mRNA expression.
Legend: Scatter plot showing the log2fold change in the RNA and protein levels as determined by RNA seq from (Philippe et al. 2020) or mass spectroscopy. Differential analysis of RNA was done using the edgeR package and the DEP (Differential Enrichment of Proteomics Analysis) Package *was used for mass spectrometry analysis. Only genes with an FDR We have not included this data in the manuscript given the deviation of the approach from our original analysis, but we are happy to reconsider the inclusion of this data to supplement our proteomic analysis.
While the LGBM modeling shows modest predictive power of sequence features alone, the manuscript stops short of exploring what cellular factors might drive context dependence. Integrating public datasets on RNA-binding protein expression or mTOR pathway activity across samples could illuminate trans-acting determinants of buffering and move beyond correlative sequence analyses,
We thank the reviewer for this suggestion. To investigate potential trans-acting determinants of buffering, we focused on 1,394 human RBPs as classified by Hentze et al. (2018), reasoning that some of these factors may facilitate translational buffering. Specifically, we examined correlations between the RNA expression of each RBP and the TE of all other genes across samples. p-values were corrected using the Bonferroni procedure. For each RBP, we then performed a Fisher’s exact test to assess whether the number of significant correlations was enriched among buffered versus non-buffered genes.
This analysis revealed that the expression levels of many RBPs are significantly enriched for either positive or negative correlations with the TE of buffered genes. In particular, we note that RNA expression of many buffered RBPs is enriched for negative correlations with the TE of other buffered transcripts. These results suggest that, rather than considering translational buffering in isolation for each transcript, buffering effects may be coordinated at the translational level and influenced by shared trans-acting factors such as RBPs. Network-based approaches have been valuable for RNA co-expression and are only now being applied to TE covariation. However, the correlative nature of these analyses limits causal inference. For example, although many ribosomal proteins appear to influence the buffering of other ribosomal proteins, they themselves may be regulated by a non-ribosomal RBP—so the apparent effects could reflect upstream regulatory influences. This analysis is now included as a supplementary figure (Sup. Fig. 5) of the revised manuscript.
Legend: A scatter plot of odds ratio log₂ of number of significant correlations (RNA abundance of RBPs ::TE of genes) and the p value from fisher test. The vertical dashed line represents the threshold odds ratio, above which RBPs exhibit a higher number of significant correlations with buffered genes. P values were corrected using Bonferroni procedure* and the horizontal dashed line represents the adjusted p value cutoff. *
Reviewer #2 (Significance (Required)):
Overall, this manuscript leverages an unprecedented compendium of matched ribosome profiling and RNAseq datasets across human cell lines and mouse tissues, combined with improved TE estimation, to robustly catalog genes exhibiting translational buffering, a clear methodological and conceptual strength. The main limitations stem from heterogeneous sample sources, largely correlative analyses, and a lack of targeted mechanistic validation. Compared to prior yeast focused studies, it fills a key gap by demonstrating conservation of buffering in mammals and linking it to dosage sensitivity and protein stability, representing a conceptual advance in understanding post-transcriptional homeostasis and a methodological step forward in TE analysis. This work will interest researchers in RNA biology, gene expression regulation, systems biology, and cancer proteomics, as well as those studying dosage-sensitive pathways and translational control. My expertise is on translational control in cancer.
We thank the reviewer for noting the broader significance of the work and for their constructive feedback.