3,295 Matching Annotations
  1. Aug 2021
    1. Alx3 decreases melanin production by directly suppressing the expression of MITF, by indirectly inhibiting the secretion of Edn3, and by indirectly promoting the expression of ASIP.

      ALX3 increases the amount of MITF.

    2. Sox10, Pax3, and Wnt3a mediated Wnt and beta-catenin signaling induce the transcription of MITF and promote differentiation of neural crest into melanoblasts XREF_BIBR - XREF_BIBR, though MITF is not expressed in the neural crest.

      WNT3A increases the amount of MITF.

    3. Inhibition of NFIB signaling in HFSCs directly stimulates expression of endothelin 2 (Edn2), which is required in HFSCs dependent McSCs activation.

      NFIB decreases the amount of EDN2.

    4. For example, MITF dependent expression of microRNA-211 promotes pigmentation in melanoblast and melanocyte cell lines by inhibiting the expression of TGF-beta receptor 2, which is involved in the maintenance of McSCs quiescence.

      Modified MITF decreases the amount of TGFBR.

    5. Alx3 decreases melanin production by directly suppressing the expression of MITF, by indirectly inhibiting the secretion of Edn3, and by indirectly promoting the expression of ASIP.

      ALX3 decreases the amount of ASIP.

    6. For example, MITF dependent expression of microRNA-211 promotes pigmentation in melanoblast and melanocyte cell lines by inhibiting the expression of TGF-beta receptor 2, which is involved in the maintenance of McSCs quiescence.

      Modified MIR211 decreases the amount of TGFB.

    7. For example, MITF dependent expression of microRNA-211 promotes pigmentation in melanoblast and melanocyte cell lines by inhibiting the expression of TGF-beta receptor 2, which is involved in the maintenance of McSCs quiescence.

      Modified MIR211 decreases the amount of TGFBR.

    8. Through interacting with PAX3, FOXD3 prevents binding of PAX3 to MITF promoter to repress melanogenesis in zebrafish, quail and chick neural crest cells XREF_BIBR, XREF_BIBR, suggesting that down-regulation of Foxd3 is a crucial step during the early phase of melanoblast lineage specification from neural crest cells.

      MITF binds PAX3.

    9. In human melanoma cells, MITF also interacts directly with beta-catenin and redirects beta-catenin transcriptional activity away from target genes regulated by Wnt and beta-catenin signaling, toward MITF specific target promoters to activate transcription 57.

      CTNNB1 binds MITF.

    10. TGF-beta binds TGF-beta receptors in melanocytes, leading to the phosphorylation of downstream effector Smad2, which inhibits melanocyte growth and melanogenesis through downregulating PAX3 and MITF transcription XREF_BIBR, XREF_BIBR.

      TGFB binds TGFBR.

    11. Notch ligands including Jagged1, Jagged2, Delta-like1, Delta-like3 and Delta-like4 bind to Notch receptor, which induces signal transduction cascade through the induction of transcription factor RBP-JK to initiate the transcription of target genes 69.

      Notch binds Notch.

    12. UV and ionizing radiation induced DNA damage triggers McSC differentiation, leading to McSC exhaustion and hair graying XREF_BIBR, XREF_BIBR, XREF_BIBR.
    13. Although alpha-MSH can derive from epithelial cells and systematically, the activation of Mc1r signaling by alpha-MSH is in the DP, which further regulates melanocyte pigmentation.

      POMC activates MC1R.

    14. These data indicate that MITF may enhance the role of Wnt and beta-catenin signaling in proliferation and differentiation of McSCs in a feedback mechanism.

      MITF activates CTNNB1.

    15. Sox10, Pax3, and Wnt3a mediated Wnt and beta-catenin signaling induce the transcription of MITF and promote differentiation of neural crest into melanoblasts XREF_BIBR - XREF_BIBR, though MITF is not expressed in the neural crest.
    16. These data indicate that MITF may enhance the role of Wnt and beta-catenin signaling in proliferation and differentiation of McSCs in a feedback mechanism.

      MITF activates Wnt.

    17. HGF, SCF, and End3 have been revealed to promote melanoblast or melanocyte proliferation and differentiation XREF_BIBR, XREF_BIBR.
    18. HGF, SCF, and End3 have been revealed to promote melanoblast or melanocyte proliferation and differentiation XREF_BIBR, XREF_BIBR.
    19. HGF, SCF, and End3 have been revealed to promote melanoblast or melanocyte proliferation and differentiation XREF_BIBR, XREF_BIBR.
    20. HGF, SCF, and End3 have been revealed to promote melanoblast or melanocyte proliferation and differentiation XREF_BIBR, XREF_BIBR.
    21. Continuous activation of beta-catenin Signaling in McSCs promotes McSCs differentiation, exhaustion and premature hair graying 8.
    22. In human melanoma cells, MITF also interacts directly with beta-catenin and redirects beta-catenin transcriptional activity away from target genes regulated by Wnt and beta-catenin signaling, toward MITF specific target promoters to activate transcription 57.
    23. Sox10, Pax3, and Wnt3a mediated Wnt and beta-catenin signaling induce the transcription of MITF and promote differentiation of neural crest into melanoblasts XREF_BIBR - XREF_BIBR, though MITF is not expressed in the neural crest.

      WNT3A activates Wnt.

    24. Inhibition of Wnt signaling by a Wnt antagonist secreted frizzled related protein 4 (sFRP4), which is exclusively expressed in the epithelial cells but not the melanocytes of the hair follicle, results in a decrease of melanocytes differentiation in the regenerating hair follicle 79.

      SFRP4 activates Wnt.

    25. PDGF promotes the proliferation of human melanoblasts and differentiation of melanocytes 104, indicating that adipose secreted PDGF may also regulate McSCs activation and differentiation.
    26. Mechanistically, knockout of Kindlin-1 promotes cutaneous epithelial stem cells differentiation via inhibiting alpha (v) beta (6) integrin mediated TGF-beta1 liberation and promoting integrin independent Wnt ligand expression to activate Wnt and beta-catenin signaling 82.

      Integrins activates TGFB1.

    27. Injecting Wnt inhibitor DKK1 into skin inhibits TPA induced the proliferation and differentiation of McSCs 52.
    28. Injecting Wnt inhibitor DKK1 into skin inhibits TPA induced the proliferation and differentiation of McSCs 52.
    29. So the white color can be due to the absence of melanocytes by non migration or death of melanocytes, or due to the suppression of differentiation by agouti or other inhibitors.

      Agouti activates cell differentiation.

    30. HGF, SCF, and End3 have been revealed to promote melanoblast or melanocyte proliferation and differentiation XREF_BIBR, XREF_BIBR.

      End3 activates cell differentiation.

    31. HGF, SCF, and End3 have been revealed to promote melanoblast or melanocyte proliferation and differentiation XREF_BIBR, XREF_BIBR.
    1. However, blocking CD32 but not CD64 to inhibit CRP induced FLS proliferation, invasiveness, and proinflammatory cytokine CXCL8 production revealed a major role for CD32 signaling in synovial inflammation, although CRP via CD64, not CD32, to induce MMP9 expression was noticed.

      CRP increases the amount of MMP9.

    2. As shown in XREF_FIG, CRP induced expression of CCL2 and IL-6 was blocked by either neutralizing antibody to CD32 or CD64 or both, suggesting that CRP signals through both CD32 and CD64 to induce expression of CCL2 and IL-6.

      CRP increases the amount of IL6.

    3. As shown in XREF_FIG, CRP induced expression of CCL2 and IL-6 was blocked by either neutralizing antibody to CD32 or CD64 or both, suggesting that CRP signals through both CD32 and CD64 to induce expression of CCL2 and IL-6.

      CRP increases the amount of CCL2.

    4. Thus, it is highly possible that high concentration of CRP in synovial fluid in patients with RA may directly bind primarily CD32 to activate p38 MAP kinase and NF-kappaB signaling in RA-FLSs to differentially regulate synovial inflammation.

      CD32 binds CRP.

    5. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.
    6. RA-FLS was a major cell type responsible for CRP production in RA patients, accounting for more than 65% of CRP producing cells as identified by co-expressing CRP and vimentin in the inflamed synovial tissues in patients with RA.
    7. As shown in XREF_FIG, multiplex cytokine assay kits assays showed that addition of CRP dose-dependently upregulated CCL2, CXCL8, IL-6, MMP2, MMP9 in RA-FLS but not in HFLS, although expression of IL-1beta and TNFalpha was not significantly changed (XREF_FIG).

      CRP activates MMP9.

    8. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.

      CRP activates MMP9.

    9. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.

      CRP activates MMP2.

    10. As shown in XREF_FIG, multiplex cytokine assay kits assays showed that addition of CRP dose-dependently upregulated CCL2, CXCL8, IL-6, MMP2, MMP9 in RA-FLS but not in HFLS, although expression of IL-1beta and TNFalpha was not significantly changed (XREF_FIG).

      CRP activates MMP2.

    11. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.

      CRP activates CXCL8.

    12. As shown in XREF_FIG, multiplex cytokine assay kits assays showed that addition of CRP dose-dependently upregulated CCL2, CXCL8, IL-6, MMP2, MMP9 in RA-FLS but not in HFLS, although expression of IL-1beta and TNFalpha was not significantly changed (XREF_FIG).

      CRP activates CXCL8.

    13. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.

      CRP activates IL6.

    14. As shown in XREF_FIG, multiplex cytokine assay kits assays showed that addition of CRP dose-dependently upregulated CCL2, CXCL8, IL-6, MMP2, MMP9 in RA-FLS but not in HFLS, although expression of IL-1beta and TNFalpha was not significantly changed (XREF_FIG).

      CRP activates IL6.

    15. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.

      CRP activates CCL2.

    16. As shown in XREF_FIG, multiplex cytokine assay kits assays showed that addition of CRP dose-dependently upregulated CCL2, CXCL8, IL-6, MMP2, MMP9 in RA-FLS but not in HFLS, although expression of IL-1beta and TNFalpha was not significantly changed (XREF_FIG).

      CRP activates CCL2.

    17. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.
    18. This was further confirmed by the ability of pre-treating RA-FLS with a NF-kappaB inhibitor, PDTC (100 mumol/L) to inhibit CRP induced proliferation (XREF_FIG) and upregulation of CXCL8, CCL2.
    19. Here we tested the hypothesis that CRP may be produced locally by FLSs and functions to induce the synovial inflammation in patients with RA.
    20. CRP may promote synovial inflammation via mechanism associated with activation of CD32/64- p38 and NF-kappaB signaling.
    21. In the present study, we found that CRP signaled primarily through CD32, to a less extent of CD64, to differentially regulate joint inflammation.
    22. CRP Promotes RA-FLS Pro inflammatory Response Differentially via the CD32/64-p 38 and NF-kappaB-Dependent Mechanisms in vitro.
    23. CRP can induce synovial inflammation via mechanisms associated with activation of CD32/64-p 38 and NF-kappaB signaling.
    24. This was supported by the findings that CRP induced activation of p38 MAP kinase and NF-kappaB signaling was blunted by neutralizing antibodies against CD32 but not CD64.

      CRP activates p38.

    25. To examine whether CRP induces NF-kappaB nuclear translation, immunofluorescence and subcellular fractionation were performed.

      CRP activates NFkappaB.

    1. We found that p-Y42 RhoA also readily translocated to the nucleus, similarly to beta-catenin, upon Wnt3A signaling (XREF_FIG A).

      RHOA translocates to the nucleus.

    2. However, the mechanism by which p-Tyr42 RhoA translocates to the nucleus remains elusive.

      RHOA translocates to the nucleus.

    3. In absence of Wnt signaling, glycogen synthase kinase-3 beta (GSK-3beta) phosphorylates beta-catenin in a ' destruction complex '.

      GSK3B phosphorylates CTNNB1.

    4. In presence of Wnt, there is the stimulation of cells in the canonical pathway as GSK-3beta phosphorylates lipoprotein receptor related protein 6 (LRP6) rather than beta-catenin, which is necessary for Axin to bind to LRP6 together with GSK-3beta and APC [XREF_BIBR].

      GSK3B phosphorylates CTNNB1.

    5. Consequently, GSK-3beta can not interact with and phosphorylate beta-catenin, resulting in increased beta-catenin stability.

      GSK3B phosphorylates CTNNB1.

    6. In particular, Tyr42 of RhoA phosphorylated by c-Src binds to IKKgamma, leading to IKKbeta activation, IkappaB phosphorylation and its degradation, resulting in NF-kappaB activation and tumorigenesis [XREF_BIBR].

      SRC phosphorylates RHOA on Y42.

    7. These results suggest that upon Wnt3A stimulation, p-Tyr216 GSK-3beta and dephospho-Ser9 GSK-3beta, both of which are known to be active forms of GSK-3beta, are critical for beta-catenin accumulation along with Tyr416 phosphorylation of Src and Tyr42 phosphorylation of RhoA (XREF_FIG C).

      SRC phosphorylates RHOA on Y42.

    8. In a previous report, we found that Tyr42 phosphorylation of RhoA (p-Tyr42 RhoA) is induced by hydrogen peroxide (H 2 O 2) in RAW264.7 cells, a macrophage cell line [XREF_BIBR].

      hydrogen peroxide leads to the phosphorylation of RHOA on Y42.

    9. Here, hydrogen peroxide also induced Tyr42 phosphorylation of Rho GTPase in HEK293T cells (XREF_FIG A).

      hydrogen peroxide leads to the phosphorylation of RHOA on Y42.

    10. Likewise, si-RhoA also prevented beta-catenin activity, and reconstitution of RhoA WT and phospho-mimetic Y42E restored the beta-catenin activity, but not with dephospho-mimetic RhoA Y42F, it having a remarkably reduced beta-catenin activity (XREF_FIG F).

      RHOA inhibits CTNNB1.

    11. However, in consistent to the previous report [XREF_BIBR], we revealed that si-RhoA impaired beta-catenin accumulation and reconstitution of RhoA restored beta-catenin accumulation (XREF_FIG A), suggesting that RhoA activity and beta-catenin stability are very closely linked, although the underlying mechanism by which RhoA induces beta-catenin accumulation in response to Wnt3A remains to be revealed.

      RHOA inhibits CTNNB1.

    12. Conversely, RhoA can be inactivated by GTPase activating proteins (GAPs) which catalyze GTP hydrolysis, leading to a GDP bound RhoA, which is inactive.

      GTP inhibits RHOA.

    13. RhoA WT and RhoA Y42E transfection to cells also had increased expression of cyclin D1 and c-Myc in presence of Wnt3A, whereas RhoA Y42F transfection inhibited this cyclin D1 and c-Myc up-regulation (XREF_FIG F).

      RHOA increases the amount of MYC.

    14. RhoA WT and RhoA Y42E transfection to cells also had increased expression of cyclin D1 and c-Myc in presence of Wnt3A, whereas RhoA Y42F transfection inhibited this cyclin D1 and c-Myc up-regulation (XREF_FIG F).

      RHOA increases the amount of Cyclin.

    15. In this study, we found that Wnt3A induces interaction of p-Tyr42 RhoA and beta-catenin and p-Tyr42 RhoA delivers beta-catenin to the nucleus, where p-Tyr42 RhoA as well as beta-catenin regulates expression of specific genes such as Vim by binding to Vim promoter.

      CTNNB1 binds RHOA.

    16. We next tried to identify whether GDP/GTP bound states of RhoA determine the interaction between RhoA and beta-catenin.

      CTNNB1 binds RHOA.

    17. In addition, it was demonstrated that p-Tyr42 RhoA directly interact with beta-catenin and nuclear localization of p-Tyr42 RhoA is required for nuclear delivery of beta-catenin.

      CTNNB1 binds RHOA.

    18. Of note, beta-catenin binds to Lef1 and activates Lef1 transcription complex, leading to down-regulation of E-cadherin expression [XREF_BIBR].

      CTNNB1 binds LEF1.

    19. ROCK2 and p-Tyr42 RhoA bind to Src, where they transmit other signaling pathway through p-p47phox (XREF_FIG H, XREF_FIG and 3L).

      SRC binds RHOA.

    20. Notably, p-Tyr42 RhoA preferentially bound to ROCK2, which in turn preferentially bound to p47phox, leading Ser345 phosphorylation of p47phox (XREF_FIG H and XREF_FIG).

      ROCK2 binds RHOA.

    21. It is notable that IKKgamma (also known as NEMO) facilitates RhoA activation via IKKgamma (NEMO) causing the dissociation of the RhoA and RhoGDI complex [XREF_BIBR].

      RhoGDI binds RHOA.

    22. Conversely, RhoA can be inactivated by GTPase activating proteins (GAPs) which catalyze GTP hydrolysis, leading to a GDP bound RhoA, which is inactive.

      GDP binds RHOA.

    23. As a small GTPase, RhoA can be activated by several guanine nucleotide exchange factors (GEFs) resulting in GTP binding to RhoA, representing an activated RhoA.

      GTP binds RHOA.

    24. Remarkably, si-RhoA impaired beta-catenin accumulation, but re-constitutive dephospho-mimetic RhoA Y42F as well as RhoA WT and phospho-mimetic RhoA Y42E allowed recovery of beta-catenin accumulation (XREF_FIG A).

      RHOA activates CTNNB1.

    25. However, in consistent to the previous report [XREF_BIBR], we revealed that si-RhoA impaired beta-catenin accumulation and reconstitution of RhoA restored beta-catenin accumulation (XREF_FIG A), suggesting that RhoA activity and beta-catenin stability are very closely linked, although the underlying mechanism by which RhoA induces beta-catenin accumulation in response to Wnt3A remains to be revealed.

      RHOA activates CTNNB1.

    26. One clue for this function is that the RhoA effector protein, ROCK2, which was supposed to be activated by p-Tyr42 RhoA (XREF_FIG H), may enhance the activation of p300 acetyltransferase by phosphorylation [XREF_BIBR].

      RHOA activates ROCK2.

    27. The results suggest that ROCK2 is a preferential downstream effector protein of p-Tyr42 RhoA while ROCK1 is activated dominantly by RhoA (XREF_FIG J).

      RHOA activates ROCK1.

    28. Consistent to the result of XREF_FIG B, si-RhoA abolished superoxide production, and reconstituted RhoA WT and phospho-mimetic RhoA Y42E restored superoxide production.

      RHOA activates superoxide.

    29. 3.4 Wnt3A induces formation of RhoA, beta, and catenin complex.

      WNT3A activates RHOA.

    30. Here, we found that Wnt3A also induces the complex formation of beta-catenin and RhoA, particularly p-Y42 RhoA.

      WNT3A activates RHOA.

    31. Wnt3A induced nuclear localization of beta-catenin (XREF_FIG A).

      WNT3A activates CTNNB1.

    32. Here, we found that Wnt3A also induces the complex formation of beta-catenin and RhoA, particularly p-Y42 RhoA.

      WNT3A activates CTNNB1.

    33. Of note, vimentin can activate RhoA through GEF-H1 (also known as ARHGEF2) [XREF_BIBR].

      VIM activates RHOA.

    34. ROS scavenger, NAC and NADPH oxidase inhibitors, DPI and apocynin abolished upregulation of p-Tyr42 Rho, beta-catenin, p-Tyr416 Src, p-Ser9 GSK-3beta and p-Tyr216 GSK-3beta upon Wnt3A in HEK293T cells, suggesting that superoxide is closely involved in the increases of the above molecules via Wnt3A (XREF_FIG C).
    35. ROS scavenger, NAC and NADPH oxidase inhibitors, DPI and apocynin abolished upregulation of p-Tyr42 Rho, beta-catenin, p-Tyr416 Src, p-Ser9 GSK-3beta and p-Tyr216 GSK-3beta upon Wnt3A in HEK293T cells, suggesting that superoxide is closely involved in the increases of the above molecules via Wnt3A (XREF_FIG C).
    36. As a small GTPase, RhoA can be activated by several guanine nucleotide exchange factors (GEFs) resulting in GTP binding to RhoA, representing an activated RhoA.

      guanine activates RHOA.

    37. Thereby, it is possible that p-Tyr42 RhoA GTPase causes the acetylation of beta-catenin through p300 acetyltransferase, remarkably enhancing beta-catenin activity as a co-activator of TCF4 in the nucleus.

      RHOA leads to the acetylation of CTNNB1.

    1. RhoA GTPase phosphorylated at tyrosine 42 by src kinase binds to beta-catenin and contributes transcriptional regulation of vimentin upon Wnt3A.

      CTNNB1 binds RHOA.

    2. Meanwhile, p-Tyr42 RhoA bound to beta-catenin via the N-terminal domain of beta-catenin, thereby leading to the nuclear translocation of p-Tyr42 RhoA, beta, and catenin complex.

      CTNNB1 binds RHOA.

    3. RhoA GTPase phosphorylated at tyrosine 42 by src kinase binds to beta-catenin and contributes transcriptional regulation of vimentin upon Wnt3A.

      RHOA activates VIM.

    4. Wnt3A also induced accumulation of beta-catenin along with activations of RhoA and ROCK1.

      WNT3A activates CTNNB1.

    1. KLK8 degrades VE-cadherin, thereby promoting plakoglobin nuclear translocation.

      KLK8 inhibits CDH5.

    2. Protein expression levels of Ki67 and PCNA were also increased by KLK8 overexpression.

      KLK8 increases the amount of MKI67.

    3. The present study found that upregulation of KLK8 leads to plakoglobin dependent nuclear translocation of p53, which binds to HIF-1alpha, and further enhances the transactivation effect of HIF-1alpha on the TGF-beta1 promoter and consequently upregulates TGF-beta1 expression at the transcriptional level.

      KLK8 increases the amount of TGFB1.

    4. A specificity protein-1 (Sp-1) consensus site was identified in the human KLK8 promoter and was found to mediate the high glucose induced KLK8 expression.

      SP1 increases the amount of KLK8.

    5. A specificity protein-1 (Sp-1) consensus site was identified in the human KLK8 promoter and was found to mediate the high glucose induced KLK8 expression.

      SP1 increases the amount of KLK8.

    6. It was found that the mRNA and protein expression levels of KLK8 were significantly upregulated in a dose dependent manner in high glucose treated HCAECs compared with those of normal glucose treated HCAECs, suggesting that high glucose stimulated KLK8 expression at the transcriptional level.

      glucose increases the amount of KLK8.

    7. To explore the molecular mechanisms involved in the high glucose induced upregulation of KLK8 expression, HCAECs were treated with increasing concentration of glucose (15 and 25 mM) for 5 days.

      glucose increases the amount of KLK8.

    8. KLK8 also induced the binding of p53 with Smad3, subsequently promoting pro EndMT reprogramming via the TGF-beta1 and Smad signaling pathway in HCAECs.

      TP53 binds SMAD3.

    9. The present study found that upregulation of KLK8 leads to plakoglobin dependent nuclear translocation of p53, which binds to HIF-1alpha, and further enhances the transactivation effect of HIF-1alpha on the TGF-beta1 promoter and consequently upregulates TGF-beta1 expression at the transcriptional level.

      TP53 binds HIF1A.

    10. While cadherin bound beta-catenin and plakoglobin are required for cell adhesion, membrane uncomplexed beta-catenin and plakoglobin have a function in transducing the Wnt signal from the cell surface to the nucleus XREF_BIBR, XREF_BIBR, XREF_BIBR.

      Cadherin binds CTNNB1.

    11. In addition, KLK8 induced p53 activation promoted the pro fibrotic reprogramming by TGF-beta1 via the cooperation of p53 with Smads in endothelial cells.

      KLK8 activates TP53.

    12. As shown in Figure XREF_FIG A, it was found that high glucose led to a significant increase in the association of plakoglobin with p53, which was blocked by KLK8 knockdown.

      KLK8 activates TP53.

    13. Taken together, these data highlight the role of KLK8 induced cooperation of p53 with Smad3 in promoting pro EndMT reprogramming by TGF-beta1 in endothelial cells.

      KLK8 activates TP53.

    14. The present study found that KLK8 overexpression significantly increased the association of p53 with Smad3, which was blocked by plakoglobin knockdown.

      KLK8 activates TP53.

    15. KLK8 overexpression induced the association of p53 with HIF-1alpha, which was blocked by plakoglobin knockdown.

      KLK8 activates TP53.

    16. Of note, since KLK8 promotes proliferation and migration in cardiac fibroblasts, a potential contribution of KLK8 induced fibroblast activation to the development of cardiac fibrosis in the context of diabetes can not be excluded.
    17. Cell proliferation assay and Ki67 immunostaining revealed that KLK8 overexpression markedly enhanced the proliferation of cardiac fibroblasts.
    18. These findings indicate that Sp-1 mediates high glucose induced upregulation of KLK8 in endothelial cells.

      SP1 activates KLK8.

    19. Sp-1 mediates high glucose induced upregulation of KLK8 in endothelial cells.

      SP1 activates KLK8.

    20. A major implication of high glucose induced KLK8 upregulation is that KLK8 targeted therapy would have clinical relevance for patients with diabetic myopathy.

      glucose activates KLK8.

    21. These findings indicate that Sp-1 mediates high glucose induced upregulation of KLK8 in endothelial cells.

      glucose activates KLK8.

    22. Sp-1 mediates high glucose induced upregulation of KLK8 in endothelial cells.

      glucose activates KLK8.

    23. KLK8 siRNA not only led to a significant decrease of KLK8 expression in HCAECs, but also blocked the high glucose induced upregulation of KLK8.

      glucose activates KLK8.

    24. High glucose may promote the plakoglobin dependent cooperation of p53 with HIF-1alpha and Smad3, subsequently increasing the expression of TGF-beta1 and the pro EndMT target genes of the TGF-beta1 and Smad signaling pathway in a KLK8 dependent manner both in vitro and in vivo.

      glucose activates TP53.

    25. Taken together, these in vitro and in vivo results suggest that high glucose may promote plakoglobin dependent cooperation of p53 with HIF-1alpha and Smad3, subsequently increasing the expression of TGF-beta1 and its pro EndMT target genes in a KLK8 dependent manner.

      glucose activates TP53.

    26. High glucose promotes plakoglobin dependent cooperation of p53 with HIF-1alpha and Smad, subsequently increasing the expression of TGF-beta1 and its pro fibrotic target genes in a KLK8 dependent manner.

      glucose activates TP53.

    27. The in vitro and in vivo findings further demonstrated that high glucose may promote plakoglobin dependent cooperation of p53 with HIF-1alpha and Smad3, subsequently increasing the expression of TGF-beta1 and the pro EndMT target genes of the TGF-beta1 and Smad signaling pathway in a KLK8 dependent manner.

      glucose activates TP53.

    28. The in vitro and in vivo findings further demonstrated that high glucose may promote plakoglobin dependent cooperation of p53 with HIF-1alpha and Smad3, subsequently increasing the expression of TGF-beta1 and the pro EndMT target genes of the TGF-beta1 and Smad signaling pathway in a KLK8 dependent manner.

      glucose activates TP53.

    1. Downregulation of PTEN expression or inhibiting its biologic activity improves heart function, promotes cardiomyocytes proliferation, reduces cardiac fibrosis as well as dilation, and inhibits apoptosis following ischemic stress such as myocardial infarction.
    1. Collectively, these data indicate that both CYP2C19 and CYP2C9 are important contributors in CBD metabolism to the active metabolite 7-OH-CBD.
    2. Collectively, these data indicate that both CYP2C19 and CYP2C9 are important contributors in CBD metabolism to the active metabolite 7-OH-CBD.
    3. Collectively, these data indicate that both CYP2C19 and CYP2C9 are important contributors in CBD metabolism to the active metabolite 7-OH-CBD.
    4. Collectively, these data indicate that both CYP2C19 and CYP2C9 are important contributors in CBD metabolism to the active metabolite 7-OH-CBD.
    1. Moreover, GAS5 and Smad4 overexpression inhibited LPS- induced chondrocytes apoptosis, while miR-146a overexpression played an opposite role and attenuated the effects of GAS5 and Smad4 overexpression on cell apoptosis.
  2. Jul 2021
    1. PTEN and PTPs all antagonize the insulin signaling as they directly interact with PI3K and IR [XREF_BIBR], and both consist of a cysteine residue in the active site that is highly susceptible to H 2 O 2 -induced oxidation.

      PTEN inhibits INS.

    2. For instance, H 2 O 2 can induce PTEN oxidation, which inactivates PTEN phosphatase function by establishing a Cys 124 -Cys 71 disulfide bond [XREF_BIBR].

      PTEN inhibits Phosphatase.

    3. For example, the Parkinson disease protein 7 (PARK7) was found to repress the PTEN phosphatase function by binding to PTEN.

      PARK7 inhibits PTEN.

    4. Recently, it has been found that the impairment of PARK2 can induce the suppression of PTEN by S nitrosylation through increase the level of NO [XREF_BIBR].

      PRKN activates PTEN.

    5. Besides, Prx II deficient MEFs induced PTEN oxidation and increased PI3K and Akt activation when exposed to insulin, which leads to an increase the insulin sensitivity.

      PRX activates PTEN.

    6. Prx III-deficiency also induces the augmentation in both PTEN oxidation and Trx dimerization.

      PRX activates PTEN.

    1. It has been demonstrated that Prx I can preserve and promote the tumor-suppressive function of PTEN by preventing oxidation of PTEN under benign oxidative stress via direct interaction.

      PRX activates PTEN.

    2. Also, Prx II deficient cells increased PTEN oxidation and insulin sensitivity.

      PRX activates PTEN.

    1. To examine whether FoxO3a is required for PTEN mediated negative regulation of autophagy, we analyzed the mRNA levels of ATG5, ATG7, and ATG12 in the ipsilateral hippocampus post-ICH.

      PTEN inhibits autophagy.

    2. In the present study, PTEN inhibition not only reduced the levels of autophagy related proteins but also activated the PI3K and AKT pathway in the ipsilateral hippocampus after ICH.

      PTEN inhibits PI3K.

    3. Specifically, PTEN antagonized the PI3K and AKT signaling and downstream effector FoxO3a phosphorylation and subsequently enhanced nuclear translocation of FoxO3a to drive proautophagy gene program, but these changes were diminished upon PTEN inhibition.

      PTEN leads to the dephosphorylation of FOXO3.

    4. Mechanistically, blockage of PTEN could enhance FoxO3a phosphorylation modification to restrict its nuclear translocation and ATG transcription via activating the PI3K and AKT pathway, leading to the suppression of the autophagic program.

      PTEN leads to the dephosphorylation of FOXO3.

    5. According to these data, we speculate that posthemorrhagic PTEN elevation triggers the nuclear accumulation of FoxO3a and subsequent transcriptional activation of ATGs, resulting in sequential activation of autophagy.

      PTEN activates FOXO3.

    6. Herein, we identified that ICH induced a significant increase in ATG transcriptional levels including ATG5, ATG7, and ATG12, which was strongly associated with PTEN mediated FoxO3a nuclear translocation.

      PTEN activates FOXO3.

    7. PTEN Inhibition Reverses Secondary Hippocampal Injury Post-ICH.
    8. Also, inactivation of the PI3K/AKT/mTOR pathway has been implicated in PTEN induced autophagy initiation [XREF_BIBR, XREF_BIBR].

      PTEN activates autophagy.

    1. S100A8 Induced Pro Inflammatory Cytokine Production Via Phosphorylation of ERK and JNK in BV-2 Cells.

      S100A8 leads to the phosphorylation of JNK.

    2. Also, the phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular-signal-regulated kinase (ERK) were stimulated by S100A8, which had an analogous effect to the lipopolysaccharide (LPS) treatment (XREF_FIG C-E).

      S100A8 leads to the phosphorylation of JNK.

    3. S100A8 Induced Pro Inflammatory Cytokine Production Via Phosphorylation of ERK and JNK in BV-2 Cells.

      S100A8 leads to the phosphorylation of ERK.

    4. Also, the phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular-signal-regulated kinase (ERK) were stimulated by S100A8, which had an analogous effect to the lipopolysaccharide (LPS) treatment (XREF_FIG C-E).

      S100A8 leads to the phosphorylation of ERK.

    5. The S100A8 knockdown using shRNA revealed that COX-2 and PGE 2 expression was regulated by S100A8, which suggested that the intracellular increase of microglial S100A8 levels upregulated COX-2 expression and PGE2 secretion, contributing to neuronal death under hypoxic conditions.

      S100A8 increases the amount of PTGS2.

    6. In agreement with previous reports, the results of this study confirmed that S100A8 significantly increased the production of IL-6, TNF-alpha, and IL-1beta.

      S100A8 activates IL6.

    7. In agreement with previous reports, the results of this study confirmed that S100A8 significantly increased the production of IL-6, TNF-alpha, and IL-1beta.

      S100A8 activates IL1B.

    8. These results suggested that S100A8, secreted by neuronal cells under hypoxic conditions, combined with TLR4 of microglia cells, activated the NLRP3 inflammasome priming.

      S100A8 activates NLRP3.

    9. These results strongly suggested that S100A8 induced the NLRP3 inflammasome priming via NF-kappaB activation.

      S100A8 activates NLRP3.

    10. The results suggested that S100A8, secreted by neuronal cells under hypoxic conditions, triggered the priming of NLRP3 in microglial cells, through the TLR4 and NF-kappaB signaling.

      S100A8 activates NLRP3.

    11. In addition, the translocation of NF-kB, which played a pivotal role in regulating the expression and activation of NLRP3, was also increased when cells were treated with S100A8.

      S100A8 activates NLRP3.

    12. In agreement with previous reports, the results of this study confirmed that S100A8 significantly increased the production of IL-6, TNF-alpha, and IL-1beta.

      S100A8 activates TNF.

    13. FACS analysis showed that the increase of S100A8 levels in microglia by hypoxia promoted neuronal apoptosis, which was confirmed by immunofluorescence.
    14. However, for the first time, we showed that up-regulation of microglial S100A8 levels increased neuronal apoptosis after hypoxia, in primary multicellular cultures consisting of neurons, astrocytes, and microglia.
    15. Therefore, this study determined whether S100A8 induced neuronal apoptosis during cerebral hypoxia and elucidated its mechanism of action using in vitro systems, including astrocytes and microglial and neuronal cells, under hypoxic conditions.
    16. S100A8 Knockdown on Microglia Attenuated Neuronal Apoptosis by Hypoxia.
    17. To investigate whether S100A8 expression in microglia induced apoptosis of neuronal cells under hypoxic condition, SH-SY5Y cells were co-cultured with BV-2 cells transfected with S100A8 shRNA for 48 h in a 0.4 mum pore transwell system and under hypoxic conditions (XREF_FIG A, B).
    18. These findings indicated that the expression of S100A8, induced in microglia cells under hypoxic conditions, activated COX-2 expression and PGE 2 secretion to induce the apoptosis of neurons.
    1. Knockdown of S100A8 levels by using shRNA revealed that microglial S100A8 expression activated COX-2 expression, leading to neuronal apoptosis under hypoxia.

      S100A8 increases the amount of PTGS2.

    2. S100A8, secreted from neurons under hypoxia, activated the secretion of tumor necrosis factor (TNF-alpha) and interleukin-6 (IL-6) through phosphorylation of extracellular-signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in microglia.

      S100A8 activates IL6.

    3. S100A8, secreted from neurons under hypoxia, activated the secretion of tumor necrosis factor (TNF-alpha) and interleukin-6 (IL-6) through phosphorylation of extracellular-signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in microglia.

      S100A8 activates TNF.

    4. The aim of this study was to determine whether S100A8 induced neuronal apoptosis during cerebral hypoxia and elucidate its mechanism of action.
    1. The precise mechanism of how LASP1 promotes PTEN ubiquitination still remains elusive 53.

      LASP1 leads to the ubiquitination of PTEN.

    2. One study showed that Nuclear Receptor Binding SET Domain Protein 2 (NSD2)-mediated dimethylation of PTEN promotes 53BP1 interactions and subsequent recruitment to sites of DNA-damage sites 75.

      NSD2 methylates PTEN.

    3. By using specific mutants of PTEN lacking lipid phosphatase function, an early study concluded that PTEN may block cell migration through a protein phosphatase mediated function on focal adhesion kinase (FAK) protein 14.

      PTEN inhibits cell migration.

    4. PTEN and PDHK1 were observed to have a synthetic-lethal relationship, as loss of PTEN and upregulation of PDHK1 in cells induced glycolysis and a dependency on PDHK1 100.
    5. This PTEN/ARID4B/PI3K signalling axis identifies a novel player in the PTEN mediated suppression of the PI3K pathway and provides a new opportunity to design novel therapeutics to target this axis to promote the tumour suppressive functions of PTEN.

      PTEN inhibits PI3K.

    6. In one of these studies, Baker et al. reported that Notch1 can mediate transcriptional suppression of PTEN, resulting in the derepression of PI3K signalling and development of trastuzumab resistance 91.

      NOTCH1 inhibits PTEN.

    7. This study was the first to link the Ras-MAPK and PI3K pathways through Notch1 transcriptional suppression of PTEN 91.

      NOTCH1 inhibits PTEN.

    8. It was reported that PTEN could dephosphorylate PGK1, a glycolytic enzyme and protein kinase with a tumorigenic role in glioblastoma 99.

      PTEN dephosphorylates PGK1.

    9. Dephosphorylation of PGK1 by PTEN was found to inhibit its activity, downstream glycolytic functions, and glioblastoma cell proliferation 99, thereby presenting another mechanism in which PTEN functions as a tumour suppressor.

      PTEN dephosphorylates PGK1.

    10. Newer studies add to this small body of data, including an intriguing study where a novel PTEN/ARID4B/PI3K pathway in which PTEN inhibits the expression of ARID4B was characterised.

      PTEN decreases the amount of ARID4B.

    11. PTEN inhibits ARID4B expression and thus prevents the transcriptional activation of ARID4B transcriptional targets PIK3CA and PIK3R2 (PI3K subunits) 79.

      PTEN decreases the amount of ARID4B.

    12. Furthermore, nuclear PTEN directly interacted with and inhibited RNA polymerase II (RNAPII)-mediated transcription, where it was involved in direct downregulation of critical transcriptional control genes including AFF4 and POL2RA 80.

      RNApo_II binds PTEN.

    13. Colocalisation of PTEN and PTENalpha promoted the function of PINK1, a mitochondrial-target kinase, and subsequently promoted energy production 105.

      PTEN activates PINK1.

    14. It is known that AKT signaling plays a critical role in the regulation of pre-mRNA splicing 77 and PTEN has been shown to modulate G6PD pre-mRNA splicing in an AKT independent manner 78.

      PTEN activates AKT.

    15. Numb inhibits Notch1, leading to the downregulation of RBP-Jkappa 94, which upregulates PTEN and anti-EMT effectors, leading to the downregulation of p-FAK and pro EMT effectors 94.

      NOTCH1 activates PTEN.

    1. Nonetheless, transcripts of genes associated with IL-17 production, such as IL17F, RORC, IL23R, and CCR6, were significantly decreased in CD8 + CD103 + CD49a + relative to CD8 + CD103 + CD49a - Trm cells, whereas transcripts for IFN-gamma were elevated (XREF_FIG D-E).

      IL23R inhibits ITGAE.

    2. Nonetheless, transcripts of genes associated with IL-17 production, such as IL17F, RORC, IL23R, and CCR6, were significantly decreased in CD8 + CD103 + CD49a + relative to CD8 + CD103 + CD49a - Trm cells, whereas transcripts for IFN-gamma were elevated (XREF_FIG D-E).

      IL23R inhibits ITGA1.

    3. CD103 binds E-cadherin, which is highly expressed on epithelia, whereas CD69 antagonizes sphingosine 1-phosphate receptor 1 (S1PR1)-mediated egress from tissues.

      CD69 inhibits S1PR1.

    4. Nonetheless, transcripts of genes associated with IL-17 production, such as IL17F, RORC, IL23R, and CCR6, were significantly decreased in CD8 + CD103 + CD49a + relative to CD8 + CD103 + CD49a - Trm cells, whereas transcripts for IFN-gamma were elevated (XREF_FIG D-E).

      IL17F inhibits ITGAE.

    5. Nonetheless, transcripts of genes associated with IL-17 production, such as IL17F, RORC, IL23R, and CCR6, were significantly decreased in CD8 + CD103 + CD49a + relative to CD8 + CD103 + CD49a - Trm cells, whereas transcripts for IFN-gamma were elevated (XREF_FIG D-E).

      IL17F inhibits ITGA1.

    6. Nonetheless, transcripts of genes associated with IL-17 production, such as IL17F, RORC, IL23R, and CCR6, were significantly decreased in CD8 + CD103 + CD49a + relative to CD8 + CD103 + CD49a - Trm cells, whereas transcripts for IFN-gamma were elevated (XREF_FIG D-E).

      CCR6 inhibits ITGAE.

    7. Nonetheless, transcripts of genes associated with IL-17 production, such as IL17F, RORC, IL23R, and CCR6, were significantly decreased in CD8 + CD103 + CD49a + relative to CD8 + CD103 + CD49a - Trm cells, whereas transcripts for IFN-gamma were elevated (XREF_FIG D-E).

      CCR6 inhibits ITGA1.

    8. Further validating transcriptional data, CXCR3 expression was higher on CD8 + CD103 + CD49a + Trm cells, whereas IL-23R and CCR6 were preferentially expressed by CD8 + CD103 + CD49a - Trm cells (XREF_FIG G).

      ITGAE increases the amount of IL23R.

    9. Further validating transcriptional data, CXCR3 expression was higher on CD8 + CD103 + CD49a + Trm cells, whereas IL-23R and CCR6 were preferentially expressed by CD8 + CD103 + CD49a - Trm cells (XREF_FIG G).

      ITGAE increases the amount of CCR6.

    10. In addition, CD8 + CD49a + Trm cells from healthy skin rapidly induced the expression of the effector molecules perforin and granzyme B when stimulated with IL-15, thereby promoting a strong cytotoxic response.

      ITGA1 increases the amount of PRF1.

    11. In addition, CD8 + CD49a + Trm cells from healthy skin rapidly induced the expression of the effector molecules perforin and granzyme B when stimulated with IL-15, thereby promoting a strong cytotoxic response.

      ITGA1 increases the amount of PRF1.

    12. In addition, CD8 + CD49a + Trm cells from healthy skin rapidly induced the expression of the effector molecules perforin and granzyme B when stimulated with IL-15, thereby promoting a strong cytotoxic response.

      ITGA1 increases the amount of GZMB.

    13. Accordingly, IL-15-dependent expression of perforin and granzyme B was augmented by IL-6, but not other cytokine combinations tested (XREF_SUPPLEMENTARY C-S2E).

      IL6 increases the amount of GZMB.

    14. Rather, their cytotoxic capacity was primed through IL-2 and IL-15-mediated induction of perforin and granzyme B expression.

      IL2 increases the amount of PRF1.

    15. Rather, their cytotoxic capacity was primed through IL-2 and IL-15-mediated induction of perforin and granzyme B expression.

      IL2 increases the amount of GZMB.

    16. In addition, CD8 + CD49a + Trm cells from healthy skin rapidly induced the expression of the effector molecules perforin and granzyme B when stimulated with IL-15, thereby promoting a strong cytotoxic response.

      CD8 increases the amount of PRF1.