S‐sulfhydration of MEK 1 leads to PARP ‐1 activation and DNA damage repair
2014; Springer Nature; Volume: 15; Issue: 7 Linguagem: Inglês
10.1002/embr.201338213
ISSN1469-3178
AutoresKexin Zhao, YoungJun Ju, Shuangshuang Li, Zaid Altaany, Rui Wang, Guangdong Yang,
Tópico(s)Genomics, phytochemicals, and oxidative stress
ResumoScientific Report5 May 2014free access Source Data S-sulfhydration of MEK1 leads to PARP-1 activation and DNA damage repair Kexin Zhao Kexin Zhao Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author YoungJun Ju YoungJun Ju Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author Shuangshuang Li Shuangshuang Li Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author Zaid Altaany Zaid Altaany Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada Department of Biology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author Rui Wang Rui Wang Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada Department of Biology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author Guangdong Yang Corresponding Author Guangdong Yang Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author Kexin Zhao Kexin Zhao Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author YoungJun Ju YoungJun Ju Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author Shuangshuang Li Shuangshuang Li Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author Zaid Altaany Zaid Altaany Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada Department of Biology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author Rui Wang Rui Wang Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada Department of Biology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author Guangdong Yang Corresponding Author Guangdong Yang Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology, Lakehead University, Thunder Bay, ON, Canada Search for more papers by this author Author Information Kexin Zhao1,2, YoungJun Ju1,2, Shuangshuang Li1,2, Zaid Altaany1,3, Rui Wang1,3 and Guangdong Yang 1,2 1Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada 2The School of Kinesiology, Lakehead University, Thunder Bay, ON, Canada 3Department of Biology, Lakehead University, Thunder Bay, ON, Canada *Corresponding author. Tel: +1 807 346 7937; Fax: +1 807 346 7873; E-mail: [email protected] EMBO Reports (2014)15:792-800https://doi.org/10.1002/embr.201338213 PDFDownload PDF of article text and main figures. Peer ReviewDownload a summary of the editorial decision process including editorial decision letters, reviewer comments and author responses to feedback. ToolsAdd to favoritesDownload CitationsTrack CitationsPermissions ShareFacebookTwitterLinked InMendeleyWechatReddit Figures & Info Abstract The repair of DNA damage is fundamental to normal cell development and replication. Hydrogen sulfide (H2S) is a novel gasotransmitter that has been reported to protect cellular aging. Here, we show that H2S attenuates DNA damage in human endothelial cells and fibroblasts by S-sulfhydrating MEK1 at cysteine 341, which leads to PARP-1 activation. H2S-induced MEK1 S-sulfhydration facilitates the translocation of phosphorylated ERK1/2 into nucleus, where it activates PARP-1 through direct interaction. Mutation of MEK1 cysteine 341 inhibits ERK phosphorylation and PARP-1 activation. In the presence of H2S, activated PARP-1 recruits XRCC1 and DNA ligase III to DNA breaks to mediate DNA damage repair, and cells are protected from senescence. Synopsis Hydrogen sulfide (H2S)—known to protect cells from aging—is shown to modify MEK1, which is necessary for ERK1/2 phosphorylation, nuclear translocation and PARP-1 activation. Thus, H2S stimulates DNA damage repair and prevents cellular senescence. H2S S-sulfhydrates and activates MEK1 at cysteine 341. H2S attenuates DNA damage in human endothelial cells via PARP-1 activation. Introduction Hydrogen sulfide (H2S) has been recognized as one of the important gasotransmitters together with nitric oxide (NO) and carbon monoxide in mammals 123. Cystathionine gamma-lyase (CSE), cystathionine beta-synthase and 3-mercaptopyruvate sulfurtransferase are the three enzymes responsible for the endogenous H2S production 2. The expressions of these enzymes are tissue-specific. Compared with all other tissues in the body, vascular tissues express abundant CSE proteins and produce a large amount of H2S 145. Deficiency of CSE reduces H2S production in vascular tissues and leads to endothelial dysfunction and high blood pressure in an age-dependent manner in mice 1. H2S has been recently characterized as both endothelium-derived relaxing factor and endothelium-derived hyperpolarizing factor 67. S-sulfhydration is proposed to mediate most of these effects from H2S by yielding a hydropersulfide moiety (–SSH) in cysteine residues of targeted proteins 8. The integrity of endothelial cell function is compromised with aging, and impaired DNA damage repair plays pivotal roles in the evolution of age-associated cardiovascular disorders 9. The salvage of DNA damage is fundamental to normal cell development and biological functions 10. DNA damage evokes a complex and highly coordinated DNA damage repair response that is integral to the maintenance of genomic stability 11. Poly(ADP-ribose)ation, carried out by poly(ADP-ribose)ation polymerases (PARPs), is one of the earliest and major cellular responses to DNA damage repair. PARPs transfer ADP-ribose from NAD+ to glutamic acid residues on a protein acceptor or itself, allowing the formation of ADP-ribose polymers (PARs) 12. Upon DNA damage, PARPs bind to DNA strand breaks and catalyze the addition of long branched chains of PARs onto itself and other chromatin remodeling factors 1213. As a result of self-poly(ADP-ribose)ation, other DNA damage repair proteins, e.g., XRCC1, pol β and DNA ligase IIIα, are recruited to the sites of DNA breaks and repair DNA damage 13. The activation of PARPs is tightly regulated by a series of kinases, in which MEK/ERK pathway plays a critical role in initiating PARP automodification 131415. We had recently demonstrated that deficiency of CSE leads to early development of cellular senescence in mouse embryonic fibroblasts (MEFs) 16. Here, we observed that H2S increases PARP-1 activity and attenuates DNA damage in human endothelial cells and fibroblasts. More DNA damage occurred in CSE-deficient mouse embryonic fibroblasts. Moreover, H2S S-sulfhydrated MEK1 at cysteine 341 and induced ERK1/2 phosphorylation, which subsequently translocates into nucleus and leads to PARP-1 activation and improved DNA damage repair and cellular senescence. Mutation of cysteine 341 in MEK1 diminished H2S-induced ERK phosphorylation and PARP-1 activation. Results and Discussion H2S induces PARP-1 activity PARP-1 is an abundant nuclear protein and plays an important role in DNA damage repair. PARP-1 activity can be reflected by PAR levels in cells or tissues 17. We first tested the impact of NaHS (a well-known H2S donor) on PARP-1 activity at lower concentration (0.1–10 μM) in human umbilical vein endothelial cells (HUVECs). NaHS treatment increased PAR level in a dose-dependent manner in HUVECs (Fig 1A). Liver and kidney are two main H2S-producing organs in mammals 818. H2S production rate was decreased approximately by 80% in both liver and kidney tissues from CSE knockout (CSE-KO) mice in comparison with wilt-type littermates (WT) 818. Consistent with the data of endothelial cells, PAR levels were lower in CSE-KO kidney and liver tissues in comparison with their counterparts from WT mice (Fig 1B and C). Murine heart tissues were reported to have lower H2S-producing potential, which is presumably due to loss of CSE translation despite the presence of CSE mRNA 19. PAR level in heart tissues from both WT and CSE-KO mice was compared, and we found that PAR is hard to detect in heart tissues from both genotypes (Fig 1D). Similar to heart tissues, PARP-1 activity in skeletal muscle and spleen tissues from both genotypes was minimal (Supplementary Fig S1A and B). Compared with liver and kidney tissues from WT mice, CSE expression in skeletal muscle and spleen tissues was pretty weaker 20. These data suggest that the level of PAR is correlated with the expression level of CSE. PAR levels in MEFs isolated from WT and CSE-KO mice were further compared, and we found significant decreased PAR levels in CSE-KO MEFs compared with WT MEFs (Fig 1E). Poly(ADP-ribose) glycohydrolase (PARG) is a major enzyme responsible for the catabolism of PAR 12. We next measured PARG activity in liver, kidney and heart tissues from both WT and CSE-KO mice. Although PARG activity was positively correlated with PAR level, the lack of CSE did not affect PARG activity in all these three tissues (Supplementary Fig S1C), suggesting H2S-stimulated PAR is not due to altered PARG activity. Higher level of PAR in kidney tissues and MEFs from WT mice observed here were unlikely due to the experimental operation with the cell exposure in the extraction buffer, because the supplement of PARP-1 inhibitor DPQ in the extraction buffer did not affect PAR level in kidney tissues and MEFs from WT mice; however, we did observe that DPQ inhibits PAR after WT MEFs are incubated with DPQ for 2 h in vitro (Supplementary Fig S2A and B). Figure 1. H2S-augmented PARP-1 activity (PAR) A. H2S-induced PAR level in HUVECs. The cells were incubated with NaHS at the indicated concentration for 2 h. *P = 0.0026; #P = 0.0002. B, C. Less PAR was found in kidney (*P = 0.031) and liver tissues (*P = 0.015) from CSE-KO mice when compared with those from WT mice. D. There was no difference in PAR level between WT heart and CSE-KO heart tissues. E. Lower PAR was detected in CSE-KO MEFs compared with WT MEFs. Data information: All the data are from at least three independent experiments. Source data are available online for this figure. Source Data for Figure 1 [embr201338213-SourceData-Fig1.pdf] Download figure Download PowerPoint Besides its role in DNA damage repair, PARP-1 has also been reported to be involved in cell apoptosis elicited by PAR-induced release of apoptosis-inducing factor (AIF) from mitochondria 21. To investigate whether H2S-stimulated PARP-1 activity induces cell apoptosis in HUVECs, we performed annexin V-FITC staining in the cells after H2S treatment. We did not observe more apoptosis (FITC positive) in HUVECs after NaHS treatment as well as cell necrosis (propidium iodide positive, Supplementary Fig S3A). In addition, no more AIF release was detected from mitochondria in HUVECs treated with NaHS (Supplementary Fig S3B). This implies that PARP-1 activation by H2S does not cause cell degeneration in HUVECs. H2S improves DNA damage repair by activating PARP-1 Considering the crucial role of PARP-1 in DNA damage repair response and the stimulatory role of H2S in PARP-1 activation observed herein, we tested the effects of H2S on DNA damage repair response. Methyl methanesulfonate (MMS), a well-known agent to induce single- and/or double-strand DNA breaks in mammalian cells, was applied here to induce DNA damage in HUVECs 22. Firstly, we compared PAR level in HUVECs treated with MMS in the presence or absence of NaHS. NaHS activated PARP-1 activity at as early as 5 min, and the supplement of MMS further strengthened NaHS-stimulated PARP-1 activity. MMS alone only activated PARP-1 activity at 30 min (Supplementary Fig S4A). To test the influence of H2S-induced PARP-1 activation on DNA damage, two methods were applied here for assessing DNA damage. The first one is apurinic/apyrimidinic (AP) contents in genomic DNA, while the other is comet tail assay 2324. More AP contents and longer comet tail would demonstrate more DNA damage inside the cells 2324. We observed more DNA damage in HUVECs treated with MMS, which was significantly reversed by NaHS co-treatment (Fig 2A and B). We further found that the administration of DPQ deteriorates H2S-restored DNA damage (Fig 2A and B) 12. This clarifies the important role of PARP-1 activation in H2S-attenuated DNA damage. Upon DNA damage, PARP-1 will recruit other DNA damage repair proteins, including XRCC1 and DNA ligase III, to repair DNA damage 13. With co-immunoprecipitation, we further found that MMS induces the binding of PARP-1 with XRCC-1 and DNA ligase III in HUVECs, which is significantly enhanced in the presence of NaHS (Fig 2C), suggesting the stimulatory role of H2S on the complex formation of PARP-1/XRCC-1/DNA ligase III. Figure 2. Mediation of PARP-1 in H2S-protected DNA damage repair H2S reduced AP contents of genomic DNA in HUVECs. *P = 0.0015; #P = 0.0028; **P = 0.0355. The data were from four independent experiments. H2S decreased comet tail length. The data were from three independent assays, and more than 30 random comet tails were calculated. *P = 9.11787E-27; #P = 2.24E-10; **P = 1.82E-09. Bar = 50 μm in all images. H2S stimulated the formation of PARP-1–XRCC-1-Ligase III complex. The data were from three independent assays. H2S reduced cellular senescence in HUVECs. For each assay, more than 4 continual fields were analyzed and 1000 cells were counted for senescence assay. *P = 6.63943E-06; #P = 4.73237E-06, **P = 0.001. Bar = 50 μm in all images. Data information: In all studies, HUVECs were treated with 1 mM MMS, 10 μM NaHS and/or 10 μM DPQ for 2 h. Source data are available online for this figure. Source Data for Figure 2 [embr201338213-SourceData-Fig2.pdf] Download figure Download PowerPoint Accumulated DNA damage is one important causative factor for cellular aging process 25. MMS treatment induced more senescent cells, which was reversed by H2S but strengthened by DPQ in HUVECs (Fig 2D). We previously reported that CSE-KO MEFs are easy to become senescent compared with WT MEFs 14. We further found here that CSE-KO MEFs have less PARP-1 activity, but more DNA damage when compared with WT MEFs even in the absence of MMS (Supplementary Fig S4B). The expression levels of CSE mRNA and protein as well as H2S production rate were significantly increased in HUVECs after MMS treatment (Supplementary Fig S4C, D and E), which probably represents one compensative response of anti-DNA damage in endothelial cells under stress condition. We further observed that NaHS stimulates PARP-1 activity and attenuates MMS-induced DNA damage in human fibroblasts (Supplementary Fig S5A and B). MMS also induced CSE expression in human fibroblasts (Supplementary Fig S5C). We previously showed that knockdown of CSE by siRNA significantly stimulates cellular senescence in human fibroblasts 16. Involvement of ERK phosphorylation in H2S-stimulated PARP-1 activity To elucidate the mechanisms underlying H2S stimulation of PARP-1 activity, we firstly tested whether H2S could directly activate PARP-1. NaHS was incubated with purified human PARP-1 for half an hour, and then PAR was detected using Western blotting. NaHS treatment did not increase PAR levels at concentration of 0.01 to 1 mM (Supplementary Fig S6A). S-sulfhydration has been identified as one novel post-translational modification by H2S in eukaryotic cells, which is the addition of one sulfhydryl to thiol side of cysteine residue and formation of persulfide group (R-S-S-H) 816. Here, we investigated whether H2S could directly S-sulfhydrate PARP-1 using biotin-switch assay. Our result showed that H2S does not S-sulfhydrate either unactivated or activated PARP-1 in HUVECs, while GAPDH is clearly S-sulfhydrated (Supplementary Fig S6B). These results indicate that H2S does not interact with PARP-1 directly. It appears that there are some other pathways for H2S-induced PARP-1 activation. It has been reported that PARP-1 can be stimulated directly by interaction with phosphorylated ERK1/2 1415. We detected the temporal change of phosphorylated ERK1/2 and PARP-1 activity in HUVECs with NaHS treatment. It was found that the levels of both PAR and phosphorylated ERK1/2 are increased by NaHS treatment (Fig 3A). The change of phosphorylated MEK1/2, which is upstream activator of ERK1/2, did not alter significantly by NaHS treatment (Fig 3A). U0126, an inhibitor of ERK1/2 phosphorylation by MEK1/2, was applied to determine the interaction between phosphorylated ERK1/2 and PARP-1 activation in HUVECs 14. The results showed U0126 decreases H2S-induced PAR (Fig 3B). All these results indicate that the involvement of MEK1/2 in H2S stimulates ERK1/2 phosphorylation and PARP-1 activity. It is worthy to note here that H2S-induced activation of ERK1/2 is not through MEK1/2 phosphorylation because NaHS has no effect on MEK1/2 phosphorylation (Fig 3A). U0126 was further applied to test the relationship between ERK phosphorylation and DNA damage in HUVECs. We observed that U0126 reverses NaHS-protected DNA damage in the presence of MMS (Fig 3C). PD98059, another inhibitor of MEK/ERK, displayed similar effects as U0126 (Supplementary Fig S7A and B) 1526. siRNA-mediated knockdown of MEK1 or ERK1/2 also prevented NaHS-induced PARP-1 activity and attenuated NaHS-protected DNA repair (Supplementary Fig S7C, D and E). All these data suggest the critical role of ERK activation in the protective effect of H2S against DNA damage. Figure 3. H2S activated PARP-1 through MEK/ERK pathway Temporal change of PARP-1 activity and phosphorylation of ERK1/2 and MEK1/2 by H2S. HUVECs were treated with 10 μM NaHS for the indicated time. p, phosphorylated; t, total. U0126 reversed H2S-stimulated PARP-1 activation in HUVECs. The cells were incubated with U0126 (10 μM) in the presence or absence of NaHS (10 μM) for 2 h. *P = 0.018 versus NaHS group. U0126 reversed H2S-protected DNA damage. HUVECs were incubated with U0126 (10 μM) in the presence or absence of MMS (1 mM) and/or NaHS (10 μM) for 2 h. *P = 5.07116E-09 versus MMS group; #P = 1.55E-09 versus MMS plus NaHS group. For each group, more than 30 random comet tails were assayed. H2S stimulated the binding of phosphorylated ERK1/2 with PARP-1. HUVECs were incubated with NaHS (10 μM) for 2 h. H2S induced phosphorylated ERK1/2 translocation into nucleus. HUVECs were incubated with NaHS (10 μM) for 2 h. *P = 0.001786; #P = 6.57978E-05. Data information: All the data are from at least three independent experiments. Source data are available online for this figure. Source Data for Figure 3 [embr201338213-SourceData-Fig3.pdf] Download figure Download PowerPoint To elucidate the involvement of MEK/ERK in the activating process of PARP-1 by H2S, we detected the interaction between ERK1/2 and PARP-1. Co-immunoprecipitaion data showed that more phosphorylated forms of ERK1/2 interact with PARP-1 after NaHS treatment (Fig 3D). Distribution of phosphorylated ERK1/2 was compared in both nuclear and cytosol part of HUVECs after NaHS treatment. We observed there is more phosphorylated ERK1/2 in nuclear fraction in NaHS-treated HUVECs compared with control cells, while similar amount of phosphorylated ERK1/2 was found in cytosols (Fig 3E). This signifies that more phosphorylated forms of ERK1/2 translocate into the nucleus and interact with PARP-1 after H2S treatment in HUVECs. H2S post-translationally modifies MEK1 via S-sulfhydration As inhibition of MEK1/2 decreases the phosphorylation of ERK1/2 and PARP-1 activity, we next investigated whether H2S could directly modify MEK1/2. After 2-h treatment with 10 μM NaHS, HUVECs were collected and subjected to biotin-switch assay. GAPDH was selected as a positive control, which is known to be S-sulfhydrated by H2S 8. We found that MEK1/2 is basically S-sulfhydrated, and NaHS treatment further increases MEK1/2 S-sulfhydration in HUVECs (Fig 4A). Classically, RAS is localized in the upstream of MEK/ERK activation cascade (RAS–RAF–MEK–ERK cascade), and RAS has been reported to be S-nitrosylated by NO at cysteine residue 118 26. Besides, ERK1/2 can be S-nitrosylated by NO as well 27. However, neither RAS nor ERK1/2 could be S-sulfhydrated after NaHS incubation (Fig 4A). And next total protein of kidney tissues from both WT and CSE-KO mice was used for biotin-switch assay as well. WT mouse kidney displayed more S-sulfhydrated MEK1/2 in comparison with that from CSE-KO mice (Supplementary Fig S8A). As MEK1 and MEK2 share higher identity for their amino acid sequences (80%), it seems reasonable to infer that both MEK1 and MEK2 can be modified by H2S. Using a MEK1- or MEK2-specific antibody, we observed that MEK1 but not MEK2 was S-sulfhydrated by NaHS in HUVECs (Fig 4B). We further observed that NaHS also S-sulfhydrates MEK1 in human fibroblasts (Supplementary Fig S8B). Figure 4. H2S S-sulfhydrated MEK1 at cysteine residue 341 and facilitated ERK phosphorylation H2S S-sulfhydrated MEK1/2 but not RAS and ERK1/2 in HUVECs. The cells were incubated with NaHS (10 μM) for 2 h and subjected to biotin-switch assay with antibodies against MEK1/2, RAS, ERK1/2 and GAPDH, respectively. H2S S-sulfhydrated MEK1 but not MEK2 in HEK293 cells. The cells were incubated with NaHS (10 μM) for 2 h and subjected to biotin-switch assay with antibodies against MEK1 and MEK2, respectively. H2S S-sulfhydrated MEK1/2 in HEK293 cells. H2S S-sulfhydrated MEK1 at cysteine residue 341. MMS enhanced MEK1 S-sulfhydration. H2S stimulated ERK2 phosphorylation in the presence of MEK1. Mutation of cysteine 341 in MEK1 diminished H2S-induced activation of ERK2. Mutation of cysteine 341 reduced H2S-induced PAR level. Data information: In (C), (D), (E) and (H), HEK293 cells were first transfected with His6-MEK1 or His6-MEK1 cysteine residue mutants for 48 h, the cells were then incubated with NaHS (10 μM) with or without MMS (1 mM) for 2 h. In (F) and (G), HEK293 cells were first transfected with His6-MEK1 or His6-MEK1 cysteine residue mutants together with GFP/ERK2 for 48 h, and the cells were then incubated with NaHS (10 μM) for 2 h. All the data were from at least three independent experiments. Source data are available online for this figure. Source Data for Figure 4 [embr201338213-SourceData-Fig4.pdf] Download figure Download PowerPoint We next constructed His6-tagged MEK1 cDNA plasmids and transfected into HEK293 cells, from which total protein was isolated and subjected to biotin-switch assay. As expected, we observed more S-sulfhydrated His6-MEK1 after NaHS treatment (Fig 4C). To investigate the molecular mechanism of MEK1 S-sulfhydration, all the six cysteine residues in MEK1 were site-mutated to glycines, including cysteine 121, 141, 207, 277, 341 and 376 28. No S-sulfhydrated His6-MEK1 was observed only when cysteine 341 was mutated, while MEK1 S-sulfhydration still existed in all other mutants (Fig 4D). MMS enhanced MEK1 S-sulfhydration in His6-MEK1-transfected cells, but failed to do so when cysteine 341 was mutated (Fig 4E). We further found that CSE overexpression induces MEK1 S-sulfhydration in HEK-293 cells (Supplementary Fig S8C). To study whether cysteine 341 in MEK1 is also responsible for H2S-stimulated ERK1/2 phosphorylation, MEK1 cDNA and GFP-ERK2 were co-transfected into HEK293 cells for 48 h 29. Then, the cells were treated with NaHS for 2 h and harvested. In control or NaHS-treated HEK293 cells only carrying GFP-ERK2, we did not observe any phosphorylation of GFP-ERK2. Thus, ERK2 is unlikely a direct target of NaHS (Fig 4F). When MEK1 was co-transfected with ERK2, phosphorylation of ERK2 was observed. Furthermore, the administration of NaHS enhanced more phosphorylated GFP-ERK2 in HEK293 cells (Fig 4F). Mutation of cysteine 341 but not cysteine 277 in MEK1 eliminated ERK2 phosphorylation even in the presence of H2S (Fig 4G). Lower PAR level was observed in HEK293 cells transfected with MEK1 C341G compared with those transfected with MEK1 C277G and wild-type MEK1 when the cells were treated with NaHS (Fig 4H). From these data, it can be postulated that cysteine 341 is critical for MEK1 activating ERK2 and PARP-1 activation. As binding of ERK2 with MEK1 is essential for the activation of ERK2 phosphorylation 30, we tested the binding affinity of MEK1, MEK1 C277G, MEK1 C341G with GFP-ERK2. MEK1 C341G displayed similar binding affinity to GFP-ERK2 as MEK1 and MEK1 C277G, which implies that cysteine 341 in MEK1 is not involved in ERK binding (Supplementary Fig S8D). H2S S-sulfhydration of cysteine 341 may elicit a conformational change of catalytic domain in MEK1, which would consequently induce the activity of MEK1. More structural investigation will be needed to address how cysteine 341 contributes to H2S-regulated MEK1 activity. In summary, we present a novel physiological aspect of H2S as a DNA damage protectant via a signal cascade, which is H2S S-sulfhydrating and activating MEK1 at cysteine 341, followed by phosphorylated ERK1/2 translocation into nucleus in stimulating PARP-1 activity through direct interaction (Fig 5). For the first time, our results clarify the role of endogenous H2S in strengthening DNA damage repair process through targeting at DNA damage repair protein PARP-1. Genetic instability due to increased DNA damage has an important impact on the pathogenesis of human diseases. Abnormal DNA damage repair interferes with fundamental cell features such as cell differentiation, cell cycle regulation and apoptosis. Deficiency of H2S is observed in various pathological conditions, including hypertension, atherosclerosis, cancers, Alzheimer's disease, gastric mucosal injury, liver cirrhosis and aging 25. The present discovery further suggests that targeting at CSE/H2S system would be a new therapeutic avenue for the treatment of DNA-damage-related diseases, and even for human life extension. Figure 5. Signal cascade of H2S stimulating PARP-1 activity and DNA damage repairPost-translational modification symbols are italicized. p, phosphorylation; SSH, S-sulfhydration; PAR, poly(ADP-ribosyl)ation. Download figure Download PowerPoint Materials and Methods Mek1 gene cloning and site mutagenesis Total RNA was isolated from HUVECs using the RNeasy mini kit (Qiagen, Hilden, Germany). One microgram of total RNA and oligo-d(T) primer was used for reverse transcriptase reaction with the AMV First Strand cDNA Synthesis kit (New England Biolabs Inc., Ipswich, MA). Digested MEK1 and pCDNA3.1/myc-his6 (Life technologies, Burlington, ON) with HindIII and BamHI were purified from agarose gel and then linked with T4 ligase (Promega, Madison, WI) for 1 h at room temperature. Single or double mutation was conducted using the QuickChange Site-Directed Mutagenesis kit (Stratagene, La Jolla, CA) 16. All primer sequences were listed in the Supplementary Table S1. Biotin-switch assay S-sulfhydration assay was performed as described previously 7816. Anti-GAPDH antibody was applied as positive control for the assay 8. Please see Supplementary Materials and Methods for detail. Western blotting, immunoprecipitation and quantitative RT-PCR See Supplementary Materials and Methods for detail. RNA interference Predesigned MEK1 and ERK1/2 siRNA were purchased from Cell Signaling technology Inc. (Beverly, MA). HUVECs were grown to more than 80% confluence and followed by siRNA transfection using transfection reagents from Santa Cruz (Dallas, TX) for 48 h prior to cell collection. AP site quantification assay Genomic DNA of HUVECs with different treatments was isolated according to the instructions of OxiSelect Oxidative DNA damage Quantification kit (Cell Biolabs, INC, San Diego, CA). Comet tail assay and senescent beta-galactosidase staining See Supplementary Materials and Methods for detail. Statistical analysis All quantitative data were all presented by mean ± standard error. Significance comparison between two groups was made with Student's t-test with two-tailed distribution. Statistical significance was determined by P-value < 0.05. Acknowledgements This study was supported by a grant-in-aid from the Heart and Stroke Foundation of Canada. G.Y. was supported by a New Investigator award from the Heart and Stroke Foundation of Canada. Author contributions KZ, RW and GY designed the study; KZ, YJ, ZA, SL and GY acquired the data; KZ and GY analyzed and interpreted the data; KZ and GY wrote and revised the manuscript. All authors discussed the results and commented on the manuscript. Conflict of interest The authors declare that they have no conflict of interest. Supporting Information Supplementary Figure S1 (application/PDF, 69.1 KB) Supplementary Figure S2 (application/PDF, 108.5 KB) Supplementary Figure S3 (application/PDF, 102.5 KB) Supplementary Figure S4 (application/PDF, 70.6 KB) Supplementary Figure S5 (application/PDF, 76.8 KB) Supplementary Figure S6 (application/PDF, 31.3 KB) Supplementary Figure S7 (application/PDF, 131.6 KB) Supplementary Figure S8 (application/PDF, 178.8 KB) Supplementary Table S1 (application/PDF, 8.2 KB) Supplementary Materials and Methods (application/PDF, 126.4 KB) Review Process File (application/PDF, 258.8 KB) References Yang G, Wu L, Jiang B, Yang W, Qi J, Cao K, Meng Q, Mustafa AK, Mu W, Zhang S et al (2008) H2S as a physiologic vasorelaxant: hypertension in mice with deletion of cystathionine gamma-lyase. Science 322: 587–590CrossrefCASPubMedWeb of Science®Google Scholar Wang R (2012) Physiological implications of hydrogen sulfide: a whiff exploration that blossomed. Physiol Rev 92: 791–896CrossrefCASPubMedWeb of Science®Google Scholar Wang R (2002) Two's company, three's a crowd: can H2S be the third endogenous gaseous transmitter? FASEB J 16: 1792–1798Wiley Online LibraryCASPubMedWeb of Science®Google Scholar Mani S, Li H, Austin RC, Dickhout JG, Lhoták S, Meng QH, Yang G, Wu L, Wang R (2013) Decreased endogenous production of hydrogen sulfide accelerates atherosclerosis. Circulation 127: 2523–2534CrossrefCASPubMedWeb of Science®Google Scholar Li L, Rose P, Moore PK (2011) Hydrogen sulfide and cell signaling. Annu Rev Pharmacol Toxicol 51: 169–187CrossrefCASPubMedWeb of Science®Google Scholar Tang G, Yang G, Jiang B, Ju Y, Wu L, Wang R (2013) H2S is an endothelium-derived hyperpolarizing factor. Antioxid Redox Signal 19: 1634–1646CrossrefCASPubMedWeb of Science®Google Scholar Mustafa AK, Sikka G, Gazi SK, Steppan J, Jung SM, Bhunia AK, Barodka VM, Gazi FK, Barrow RK, Wang R et al (2011) Hydrogen sulfide as endothelium-derived hyperpolarizing factor sulfhydrates potassium channels. Circ Res 109: 1259–1268CrossrefCASPubMedWeb of Science®Google Scholar Mustafa AK, Gadalla MM, Sen N, Kim S, Mu W, Gazi SK, Barrow RK, Yang G, Wang R, Snyder SH (2009) H2S signals through protein S-sulfhydration. Sci Signal 2: ra72CrossrefPubMedWeb of Science®Google Scholar Shirodkar AV, St Bernard R, Gavryushova A, Kop A, Knight BJ, Yan MS, Man HS, Sud M, Hebbel RP, Oettgen P et al (2013) A mechanistic role for DNA methylation in endothelial cell (EC)-enriched gene expression: relationship with DNA replication timing. Blood 121: 3531–3540CrossrefCASPubMedWeb of Science®Google Scholar Cuozzo C, Porcellini A, Angrisano T, Morano A, Lee B, Di Pardo A, Messina S, Iuliano R, Fusco A, Santillo MR et al (2007) DNA damage, homology-directed repair, and DNA methylation. PLoS Genet 3: e110CrossrefCASPubMedWeb of Science®Google Scholar D'Amours D, Desnoyers S, D'Silva I, Poirier GG (1999) Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions. Biochem J 342: 249–268CrossrefCASPubMedWeb of Science®Google Scholar Shilovsky GA, Khokhlov AN, Shram SI (2013) The protein poly(ADP-ribosyl)ation system: its role in genome stability and lifespan determination. Biochemistry 78: 433–444CASWeb of Science®Google Scholar Abdallah Y, Gligorievski D, Kasseckert SA, Dieterich L, Schäfer M, Kuhlmann CR, Noll T, Sauer H, Piper HM, Schäfer C (2007) The role of poly(ADP-ribose) polymerase (PARP) in the autonomous proliferative response of endothelial cells to hypoxia. Cardiovasc Res 73: 568–574CrossrefCASPubMedWeb of Science®Google Scholar Cohen-Armon M, Visochek L, Rozensal D, Kalal A, Geistrikh I, Klein R, Bendetz-Nezer S, Yao Z, Seger R (2007) DNA-independent PARP-1 activation by phosphorylated ERK2 increases Elk1 activity: a link to histone acetylation. Mol Cell 25: 297–308CrossrefCASPubMedWeb of Science®Google Scholar Kauppinen TM, Chan WY, Suh SW, Wiggins AK, Huang EJ, Swanson RA (2006) Direct phosphorylation and regulation of poly(ADP-ribose) polymerase-1 by extracellular signal-regulated kinases 1/2. Proc Natl Acad Sci USA 103: 7136–7141CrossrefCASPubMedWeb of Science®Google Scholar Yang G, Zhao K, Ju Y, Mani S, Cao Q, Puukila S, Khaper N, Wu L, Wang R (2013) Hydrogen sulfide protects against cellular senescence via S-sulfhydration of Keap1 and activation of Nrf2. Antioxid Redox Signal 18: 1906–1919CrossrefCASPubMedWeb of Science®Google Scholar Affar EB, Duriez PJ, Shah RG, Sallmann FR, Bourassa S, Kupper JH, Burkle A, Poirier GG (1998) Immunodot blot method for the detection of poly(ADP-ribose) synthesized in vitro and in vivo. Anal Biochem 259: 280–283CrossrefCASPubMedWeb of Science®Google Scholar Bos EM, Wang R, Snijder PM, Boersema M, Damman J, Fu M, Moser J, Hillebrands JL, Ploeg RJ, Yang G et al (2013) Cystathionine γ-lase protects against renal ischemia/reperfusion by modulating oxidative stress. J Am Soc Nephrol 24: 759–770CrossrefCASPubMedWeb of Science®Google Scholar Fu M, Zhang W, Yang G, Wang R (2012) Is cystathionine gamma-lyase protein expressed in the heart? Biochem Biophys Res Commun 428: 469–474CrossrefCASPubMedWeb of Science®Google Scholar Ishii I, Akahoshi N, Yu XN, Kobayashi Y, Namekata K, Komaki G, Kimura H (2004) Murine cystathionine gamma-lyase: complete cDNA and genomic sequences, promoter activity, tissue distribution and developmental expression. Biochem J 381: 113–123CrossrefCASPubMedWeb of Science®Google Scholar Xu H, Luo P, Zhao Y, Zhao M, Yang Y, Chen T, Huo K, Han H, Fei Z (2013) Iduna protects HT22 cells from hydrogen peroxide-induced oxidative stress through interfering poly(ADP-ribose) polymerase-1-induced cell death (parthanatos). Cell Signal 25: 1018–1026CrossrefCASPubMedWeb of Science®Google Scholar Song CX, He C (2013) Potential functional roles of DNA demethylation intermediates. Trends Biochem Sci 38: 480–484CrossrefCASPubMedWeb of Science®Google Scholar Wang Y, Liu L, Wu C, Bulgar A, Somoza E, Zhu W, Gerson SL (2009) Direct detection and quantification of abasic sites for in vivo studies of DNA damage and repair. Nucl Med Biol 36: 975–983CrossrefCASWeb of Science®Google Scholar Schlörmann W, Glei M (2012) Detection of DNA damage by comet fluorescence in situ hybridization. Methods Mol Biol 920: 91–100CrossrefCASGoogle Scholar Durik M, Kavousi M, van der Pluijm I, Isaacs A, Cheng C, Verdonk K, Loot AE, Oeseburg H, Bhaggoe UM, Leijten F et al (2012) Nucleotide excision DNA repair is associated with age-related vascular dysfunction. Circulation 126: 468–478CrossrefCASPubMedWeb of Science®Google Scholar Dai Y, Chen S, Pei XY, Almenara JA, Kramer LB, Venditti CA, Dent P, Grant S (2008) Interruption of the Ras/MEK/ERK signaling cascade enhances Chk1 inhibitor-induced DNA damage in vitro and in vivo in human multiple myeloma cells. Blood 112: 2439–2449CrossrefCASPubMedWeb of Science®Google Scholar Feng X, Sun T, Bei Y, Ding S, Zheng W, Lu Y, Shen P (2013) S-nitrosylation of ERK inhibits ERK phosphorylation and induces apoptosis. Sci Rep 3: 1814CrossrefCASPubMedWeb of Science®Google Scholar Schwartz MA, Madhani HD (2006) Control of MAPK signaling specificity by a conserved residue in the MEK-binding domain of the yeast scaffold protein Ste5. Curr Genet 49: 351–363CrossrefCASPubMedWeb of Science®Google Scholar Rubinfeld H, Hanoch T, Seger R (1999) Identification of a cytoplasmic-retention sequence in ERK2. J Biol Chem 274: 30349–30352CrossrefCASPubMedWeb of Science®Google Scholar Robinson FL, Whitehurst AW, Raman M, Cobb MH (2002) Identification of novel point mutations in ERK2 that selectively disrupt binding to MEK1. J Biol Chem 277: 14844–14852CrossrefCASPubMedWeb of Science®Google Scholar Previous ArticleNext Article Read MoreAbout the coverClose modalView large imageVolume 15,Issue 7,July 2014Cover picture: Inspired by the Scientific Report p 758 | Cover image provided by the authors. Illustration by René Pascal, Center of Advanced European Studies and Research, Department of Molecular Sensory System. Volume 15Issue 71 July 2014In this issue FiguresReferencesRelatedDetailsLoading ...
Referência(s)