Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Hydrogen sulfide anion regulates redox signaling via electrophile sulfhydration

Abstract

An emerging aspect of redox signaling is the pathway mediated by electrophilic byproducts, such as nitrated cyclic nucleotide (for example, 8-nitroguanosine 3′,5′-cyclic monophosphate (8-nitro-cGMP)) and nitro or keto derivatives of unsaturated fatty acids, generated via reactions of inflammation-related enzymes, reactive oxygen species, nitric oxide and secondary products. Here we report that enzymatically generated hydrogen sulfide anion (HS) regulates the metabolism and signaling actions of various electrophiles. HS reacts with electrophiles, best represented by 8-nitro-cGMP, via direct sulfhydration and modulates cellular redox signaling. The relevance of this reaction is reinforced by the significant 8-nitro-cGMP formation in mouse cardiac tissue after myocardial infarction that is modulated by alterations in HS biosynthesis. Cardiac HS, in turn, suppresses electrophile-mediated H-Ras activation and cardiac cell senescence, contributing to the beneficial effects of HS on myocardial infarction–associated heart failure. Thus, this study reveals HS-induced electrophile sulfhydration as a unique mechanism for regulating electrophile-mediated redox signaling.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Regulation of 8-nitro-cGMP–induced protein S-guanylation by HS-producing enzymes via sulfhydration of 8-nitro-cGMP by HS.
Figure 2: Electrophile sulfhydration by HS.
Figure 3: Cellular formation of 8-SH-cGMP and its metabolic fate.
Figure 4: Cellular HS formation and its physiological relevance to electrophile metabolism.
Figure 5: Electrophilic H-Ras activation regulated by HS in cells and in vivo in cardiac tissues.
Figure 6: HS regulation of H-Ras electrophile sensing and signaling.
Figure 7: 8-Nitro-cGMP–induced H-Ras activation in cardiac cells and their suppression by HS.
Figure 8: Dissociation of H-Ras from rafts and its activation induced by H-Ras Cys184 S-guanylation.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. Kajimura, M., Fukuda, R., Bateman, R.M., Yamamoto, T. & Suematsu, M. Interactions of multiple gas-transducing systems: hallmarks and uncertainties of CO, NO, and H2S gas biology. Antioxid. Redox Signal. 13, 157–192 (2010).

    Article  CAS  Google Scholar 

  2. Li, L., Rose, P. & Moore, P.K. Hydrogen sulfide and cell signaling. Annu. Rev. Pharmacol. Toxicol. 51, 169–187 (2011).

    Article  CAS  Google Scholar 

  3. Fridovich, I. The biology of oxygen radicals. Science 201, 875–880 (1978).

    Article  CAS  Google Scholar 

  4. Sawa, T. et al. Protein S-guanylation by the biological signal 8-nitroguanosine 3′,5′-cyclic monophosphate. Nat. Chem. Biol. 3, 727–735 (2007).

    Article  CAS  Google Scholar 

  5. Rhee, S.G. Cell signaling. H2O2, a necessary evil for cell signaling. Science 312, 1882–1883 (2006).

    Article  Google Scholar 

  6. D′Autréaux, B. & Toledano, M.B. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 8, 813–824 (2007).

    Article  Google Scholar 

  7. Akaike, T., Fujii, S., Sawa, T. & Ihara, H. Cell signaling mediated by nitrated cyclic guanine nucleotide. Nitric Oxide 23, 166–174 (2010).

    Article  CAS  Google Scholar 

  8. Rudolph, T.K. & Freeman, B.A. Transduction of redox signaling by electrophile-protein reactions. Sci. Signal. 2, re7 (2009).

    Article  Google Scholar 

  9. Forman, H.J., Fukuto, J.M. & Torres, M. Redox signaling: thiol chemistry defines which reactive oxygen and nitrogen species can act as second messengers. Am. J. Physiol. Cell Physiol. 287, C246–C256 (2004).

    Article  CAS  Google Scholar 

  10. Zaki, M.H. et al. Cytoprotective function of heme oxygenase 1 induced by a nitrated cyclic nucleotide formed during murine salmonellosis. J. Immunol. 182, 3746–3756 (2009).

    Article  CAS  Google Scholar 

  11. Uchida, K. & Shibata, T. 15-Deoxy-Δ12,14-prostaglandin J2: an electrophilic trigger of cellular responses. Chem. Res. Toxicol. 21, 138–144 (2008).

    Article  Google Scholar 

  12. Fujii, S. et al. The critical role of nitric oxide signaling, via protein S-guanylation and nitrated cyclic GMP, in the antioxidant adaptive response. J. Biol. Chem. 285, 23970–23984 (2010).

    Article  CAS  Google Scholar 

  13. Hughes, M.N., Centelles, M.N. & Moore, K.P. Making and working with hydrogen sulfide: the chemistry and generation of hydrogen sulfide in vitro and its measurement in vivo: a review. Free Radic. Biol. Med. 47, 1346–1353 (2009).

    Article  CAS  Google Scholar 

  14. Tiranti, V. et al. Loss of ETHE1, a mitochondrial dioxygenase, causes fatal sulfide toxicity in ethylmalonic encephalopathy. Nat. Med. 15, 200–205 (2009).

    Article  CAS  Google Scholar 

  15. Wintner, E.A. et al. A monobromobimane-based assay to measure the pharmacokinetic profile of reactive sulphide species in blood. Br. J. Pharmacol. 160, 941–957 (2010).

    Article  CAS  Google Scholar 

  16. Winterbourn, C.C. & Metodiewa, D. Reactivity of biologically important thiol compounds with superoxide and hydrogen peroxide. Free Radic. Biol. Med. 27, 322–328 (1999).

    Article  CAS  Google Scholar 

  17. Glushchenko, A.V. & Jocobsen, D.W. Molecular targeting of proteins by L-homocysteine: mechanistic implications for vascular disease. Antioxid. Redox Signal. 9, 1883–1898 (2007).

    Article  CAS  Google Scholar 

  18. LoPachin, R.M., Gavin, T., Geohagen, B.C. & Das, S. Neurotoxic mechanisms of electrophilic type-2 alkenes: soft-soft interactions described by quantum mechanical parameters. Toxicol. Sci. 98, 561–570 (2007).

    Article  CAS  Google Scholar 

  19. Ishii, I. et al. Murine cystathionine γ-lyase: complete cDNA and genomic sequences, promoter activity, tissue distribution and developmental expression. Biochem. J. 381, 113–123 (2004).

    Article  CAS  Google Scholar 

  20. Lander, H.M. et al. Redox regulation of cell signalling. Nature 381, 380–381 (1996).

    Article  CAS  Google Scholar 

  21. Oliva, J.L. et al. The cyclopentenone 15-deoxy-Δ12,14-prostaglandin J2 binds to and activates H-Ras. Proc. Natl. Acad. Sci. USA 100, 4772–4777 (2003).

    Article  CAS  Google Scholar 

  22. Serrano, M., Lin, A.W., McCurrach, M.E., Beach, D. & Lowe, S.W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 (1997).

    Article  CAS  Google Scholar 

  23. Wu, C., Miloslavskaya, I., Demontis, S., Maestro, R. & Galaktionov, K. Regulation of cellular response to oncogenic and oxidative stress by Seladin-1. Nature 432, 640–645 (2004).

    Article  CAS  Google Scholar 

  24. Shih, H., Lee, B., Lee, R.J. & Boyle, A.J. The aging heart and post-infarction left ventricular remodeling. J. Am. Coll. Cardiol. 57, 9–17 (2011).

    Article  Google Scholar 

  25. Liu, Y.H. et al. Role of inducible nitric oxide synthase in cardiac function and remodeling in mice with heart failure due to myocardial infarction. Am. J. Physiol. Heart Circ. Physiol. 289, H2616–H2623 (2005).

    Article  CAS  Google Scholar 

  26. Zhang, P. et al. Inducible nitric oxide synthase deficiency protects the heart from systolic overload-induced ventricular hypertrophy and congestive heart failure. Circ. Res. 100, 1089–1098 (2007).

    Article  CAS  Google Scholar 

  27. Gelb, B.D. & Tartaglia, M. Ras signaling pathway mutations and hypertrophic cardiomyopathy: getting into and out of the thick of it. J. Clin. Invest. 121, 844–847 (2011).

    Article  CAS  Google Scholar 

  28. Sano, M. et al. p53-induced inhibition of Hif-1 causes cardiac dysfunction during pressure overload. Nature 446, 444–448 (2007).

    Article  CAS  Google Scholar 

  29. Hunter, J.J., Tanaka, N., Rockman, H.A., Ross, J. Jr. & Chien, K.R. Ventricular expression of a MLC-2v-ras fusion gene induces cardiac hypertrophy and selective diastolic dysfunction in transgenic mice. J. Biol. Chem. 270, 23173–23178 (1995).

    Article  CAS  Google Scholar 

  30. Asano, K. et al. Constitutive and inducible nitric oxide synthase gene expression, regulation, and activity in human lung epithelial cells. Proc. Natl. Acad. Sci. USA 91, 10089–10093 (1994).

    Article  CAS  Google Scholar 

  31. Pechkovsky, D.V. et al. Pattern of NOS2 and NOS3 mRNA expression in human A549 cells and primary cultured AEC II. Am. J. Physiol. Lung Cell Mol. Physiol. 282, L684–L692 (2002).

    Article  CAS  Google Scholar 

  32. Nishida, M. et al. Heterologous down-regulation of angiotensin type 1 receptors by purinergic P2Y2 receptor stimulation through S-nitrosylation of NF-κB. Proc. Natl. Acad. Sci. USA 108, 6662–6667 (2011).

    Article  CAS  Google Scholar 

  33. Hancock, J.F. Ras proteins: different signals from different locations. Nat. Rev. Mol. Cell Biol. 4, 373–384 (2003).

    Article  CAS  Google Scholar 

  34. Prior, I.A. et al. GTP-dependent segregation of H-Ras from lipid rafts is required for biological activity. Nat. Cell Biol. 3, 368–375 (2001).

    Article  CAS  Google Scholar 

  35. Roy, S. et al. Individual palmitoyl residues serve distinct roles in H-Ras trafficking, microlocalization, and signaling. Mol. Cell. Biol. 25, 6722–6733 (2005).

    Article  CAS  Google Scholar 

  36. Carballal, S. et al. Reactivity of hydrogen sulfide with peroxynitrite and other oxidants of biological interest. Free Radic. Biol. Med. 50, 196–205 (2011).

    Article  CAS  Google Scholar 

  37. Zhao, W., Zhang, J., Lu, Y. & Wang, R. The vasorelaxant effect of H2S as a novel endogenous gaseous KATP channel opener. EMBO J. 20, 6008–6016 (2001).

    Article  CAS  Google Scholar 

  38. Bucci, M. et al. Hydrogen sulfide is an endogenous inhibitor of phosphodiesterase activity. Arterioscler. Thromb. Vasc. Biol. 30, 1998–2004 (2010).

    Article  CAS  Google Scholar 

  39. Sun, P. et al. PRAK is essential for ras-induced senescence and tumor suppression. Cell 128, 295–308 (2007).

    Article  CAS  Google Scholar 

  40. Yoshitake, J. et al. Nitric oxide as an endogenous mutagen for Sendai virus without antiviral activity. J. Virol. 78, 8709–8719 (2004).

    Article  CAS  Google Scholar 

  41. Ohshima, H., Sawa, T. & Akaike, T. 8-Nitroguanine, a product of nitrative DNA damage caused by reactive nitrogen species: formation, occurrence, and implications in inflammation and carcinogenesis. Antioxid. Redox Signal. 8, 1033–1045 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J.B. Gandy for her editing of the manuscript. Thanks are also due to T. Okamoto, S. Fujii, Md. M. Rahaman, S. Khan, K.A. Ahmed, J. Yoshitake, T. Matsunaga, M.H.A. Rahman, J. Sakamoto, J. Minkyung, K. Hara, M. Goto, F. Sohma, K. Taguchi, T. Miura, T. Toyama, Y. Shinkai, I. Ishii and M. Toyataka for technical assistance; S. Kasamatsu, T. Ida and K. Kunieda for 8-nitro-cGMP preparation; Y. Kawai for technical assistance with LC/MS/MS experiments; H. Arimoto for helpful discussion; and J. Wu for reading our paper to evaluate its concepts and interdisciplinary accessibility. This work was supported in part by Grants-in-Aid for Scientific Research and Grants-in-Aid for Scientific Research on Innovative Areas (Research in a Proposed Area) from the Ministry of Education, Sciences, Sports and Technology, Japan; a grant from the Japan Science and Technology Agency PRESTO program; grants from the Ministry of Health, Labor and Welfare of Japan; and grants from the US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

M.N., T.A. and T. Sawa designed the experiment, performed analysis and wrote the paper; N.K., H. Inoue and T. Shibata performed MS analysis, cell biology and mouse and rat studies; K.O., H. Ihara, H.M., H.K., M.S. and M.Y. performed data analyses; B.A.F., A.v.d.V., K.U. and Y.K. designed the experiment and edited the paper.

Corresponding author

Correspondence to Takaaki Akaike.

Ethics declarations

Competing interests

B.A.F. acknowledges financial interests in Complexa, Inc.

Supplementary information

Supplementary Text and Figures

Supplementary Methods and Supplementary Results (PDF 9048 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nishida, M., Sawa, T., Kitajima, N. et al. Hydrogen sulfide anion regulates redox signaling via electrophile sulfhydration. Nat Chem Biol 8, 714–724 (2012). https://doi.org/10.1038/nchembio.1018

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.1018

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing