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.

  • Review Article
  • Published:

Protein S-nitrosylation: purview and parameters

Key Points

  • Nitric oxide (NO) is produced enzymatically in most or all cell types and tissues. The modification by NO of prosthetic metals in proteins — in particular, haem iron — was shown to mediate some important effects of NO. Subsequently, it emerged that the addition of an NO group to the thiol side chain of cysteine residues within proteins and peptides, which is designated S-nitrosylation, conveys a large part of the ubiquitous influence of NO on cellular signal transduction.

  • Proteins in most or all functional classes function as substrates for S-nitrosylation in vitro and in vivo, and a growing body of research shows the occurrence and effects of endogenous S-nitrosylation in intact cellular systems.

  • It has become clear that S-nitrosylation and de-nitrosylation are precisely regulated in space and time. The specificity of S-nitrosylation within and between proteins is conferred by structural motifs and allosteric regulators, as well as by interactions between NO synthases and target proteins, which might themselves be modulated by S-nitrosylation. Enzymatic activities that promote S-nitrosylation and de-nitrosylation have been identified, but the mechanisms of dynamic regulation in situ remain largely unexplored.

  • Recent work has revealed new effector mechanisms for S-nitrosylation, including the regulation of protein–protein interactions, subcellular localization of proteins and ubiquitylation-dependent protein degradation

  • S-nitrosylation regulates cellular mechanisms that underlie a wide range of critical functions including apoptosis, cellular metabolism, membrane trafficking, protein phosphorylation, the activity of enzymes through both allosteric and active-site modification, transcription-factor stability and activity, receptor-coupled and other ion-channel activity, and maintenance of cellular redox equilibrium (responses to oxidative and nitrosative stress).

  • The elucidation of the physiological roles of S-nitrosylation has begun to impact on the understanding of human health and disease, and the dysregulation of S-nitrosylation is associated with a growing list of pathophysiological conditions (endotoxic shock, multiple sclerosis, Parkinson's disease, pulmonary hypertension, sickle cell disease and asthma).

Abstract

S-nitrosylation, the covalent attachment of a nitrogen monoxide group to the thiol side chain of cysteine, has emerged as an important mechanism for dynamic, post-translational regulation of most or all main classes of protein. S-nitrosylation thereby conveys a large part of the ubiquitous influence of nitric oxide (NO) on cellular signal transduction, and provides a mechanism for redox-based physiological regulation.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Cellular sources and targets of nitric oxide.
Figure 2: Concerted acid–base catalysis of protein transnitrosylation.
Figure 3: Regulation by S-nitrosylation of protein–protein interactions of caspase-3.
Figure 4: Regulation of apoptosis through TRX–ASK1 and preservation of the redox equilibrium.
Figure 5: Regulation of signal transduction through Src by S-nitrosylation at multiple loci.
Figure 6: Regulation by S-nitrosylation of protein ubiquitylation and proteasomal targeting.

Similar content being viewed by others

References

  1. Murad, F. Cyclic guanosine monophosphate as a mediator of vasodilation. J. Clin. Invest. 78, 1–5 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bredt, D. S. Endogenous nitric oxide synthesis: biological functions and pathophysiology. Free Radic. Res. 31, 577–596 (1999).

    Article  CAS  PubMed  Google Scholar 

  3. Stamler, J. S. et al. S-nitrosylation of proteins with nitric oxide: synthesis and characterization of biologically active compounds. Proc. Natl Acad. Sci. USA 89, 444–448 (1992). The first demonstration of protein S -nitrosylation, which showed that proteins in several classes could be modified at active-site or allosteric Cys residues by endogenous and exogenous NO.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Stamler, J. S. et al. S-nitrosylation of tissue-type plasminogen activator confers vasodilatory and antiplatelet properties on the enzyme. Proc. Natl Acad. Sci. USA 89, 8087–8091 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Stamler, J. S. et al. in Biology of Nitric Oxide (eds, Moncada, S., Marletta, M. A. & Hibbs, J. B. J.) 20–23 (Portland Press, London, UK, 1992).

    Google Scholar 

  6. Stamler, J. S., Lamas, S. & Fang, F. C. Nitrosylation: the prototypic redox-based signaling mechanism. Cell 106, 675–683 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Jaffrey, S. R., Erdjument-Bromage, H., Ferris, C. D., Tempst, P. & Snyder, S. H. Protein S-nitrosylation: a physiological signal for neuronal nitric oxide. Nature Cell Biol. 3, 193–197 (2001). Introduced a method to selectively biotinylate sites of S -nitrosylation within proteins, and applied this method to reveal several endogenous substrates in neural tissue of nNOS-dependent S -nitrosylation.

    Article  CAS  PubMed  Google Scholar 

  8. Liu, L. et al. Essential roles of S-nitrosothiols in vascular homeostasis and endotoxic shock. Cell 116, 617–628 (2004). Found that mice with a targeted gene deletion of GSNO reductase have large increases in tissue damage and mortality following endotoxic challenge, which indicated an important role for S -nitrosothiol metabolism in innate immunity, as well as effects on basal SNO levels and vascular tone.

    Article  CAS  PubMed  Google Scholar 

  9. de Jesus-Berrios, M. et al. Enzymes that counteract nitrosative stress promote fungal virulence. Curr. Biol. 13, 1963–1968 (2003).

    Article  CAS  PubMed  Google Scholar 

  10. Foster, M. W., McMahon, T. J. & Stamler, J. S. S-nitrosylation in health and disease. Trends Mol. Med. 9, 160–168 (2003).

    Article  CAS  PubMed  Google Scholar 

  11. Lane, P., Hao, G. & Gross, S. S. S-nitrosylation is emerging as a specific and fundamental posttranslational protein modification: head-to-head comparison with O-phosphorylation. Sci. STKE, RE1 (2001).

  12. Boehning, D. & Snyder, S. H. Novel neural modulators. Annu. Rev. Neurosci. 26, 105–131 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Barouch, L. A. et al. Nitric oxide regulates the heart by spatial confinement of nitric oxide synthase isoforms. Nature 416, 337–339 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Gow, A. J. et al. Basal and stimulated protein S-nitrosylation in multiple cell types and tissues. J. Biol. Chem. 277, 9637–9640 (2002).

    Article  CAS  PubMed  Google Scholar 

  15. Stamler, J. S. & Toone, E. J. The decomposition of thionitrites. Curr. Opin. Chem. Biol. 6, 779–785 (2002).

    Article  CAS  PubMed  Google Scholar 

  16. Bartberger, M. D. et al. S–N dissociation energies of S-nitrosothiols: on the origins of nitrosothiol decomposition rates. J. Am. Chem. Soc. 123, 8868–8869 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Stamler, J. S. S-nitrosothiols in the blood: roles, amounts, and methods of analysis. Circ. Res. 94, 414–417 (2004).

    Article  CAS  PubMed  Google Scholar 

  18. Ckless, K. et al. In situ detection and visualization of S-nitrosylated proteins following chemical derivatization: identification of Ran GTPase as a target for S-nitrosylation. Nitric Oxide 11, 216–217 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Mannick, J. B. et al. Fas-induced caspase denitrosylation. Science 284, 651–654 (1999). Provided the first demonstration of stimulus-coupled protein de-nitrosylation, which was triggered by activation of a membrane receptor that subserves apoptotic stimulation.

    Article  CAS  PubMed  Google Scholar 

  20. Hoffmann, J., Haendeler, J., Zeiher, A. M. & Dimmeler, S. TNF α and oxLDL reduce protein S-nitrosylation in endothelial cells. J. Biol. Chem. 276, 41383–41387 (2001).

    Article  CAS  PubMed  Google Scholar 

  21. Arnelle, D. R. & Stamler, J. S. NO+, NO, and NO donation by S-nitrosothiols: implications for regulation of physiological functions by S-nitrosylation and acceleration of disulfide formation. Arch. Biochem. Biophys. 318, 279–285 (1995).

    Article  CAS  PubMed  Google Scholar 

  22. Gow, A. J., Luchsinger, B. P., Pawloski, J. R., Singel, D. J. & Stamler, J. S. The oxyhemoglobin reaction of nitric oxide. Proc. Natl Acad. Sci. USA 96, 9027–9032 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Romeo, A. A., Capobianco, J. A. & English, A. M. Superoxide dismutase targets NO from GSNO to Cysβ93 of oxyhemoglobin in concentrated but not dilute solutions of the protein. J. Am. Chem. Soc. 125, 14370–14378 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Mani, K., Cheng, F., Havsmark, B., David, S. & Fransson, L. A. Involvement of glycosylphosphatidylinositol-linked ceruloplasmin in the copper/zinc-nitric oxide-dependent degradation of glypican-1 heparan sulfate in rat C6 glioma cells. J. Biol. Chem. 279, 12918–12923 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Inoue, K. et al. Nitrosothiol formation catalyzed by ceruloplasmin. Implication for cytoprotective mechanism in vivo. J. Biol. Chem. 274, 27069–27075 (1999).

    Article  CAS  PubMed  Google Scholar 

  26. Stubauer, G., Giuffre, A. & Sarti, P. Mechanism of S-nitrosothiol formation and degradation mediated by copper ions. J. Biol. Chem. 274, 28128–28133 (1999).

    Article  CAS  PubMed  Google Scholar 

  27. Tao, L. & English, A. M. Mechanism of S-nitrosation of recombinant human brain calbindin D28K. Biochemistry 42, 3326–3334 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. Romeo, A. A., Capobianco, J. A. & English, A. M. Heme nitrosylation of deoxyhemoglobin by S-nitrosoglutathione requires copper. J. Biol. Chem. 277, 24135–24141 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Luchsinger, B. P. et al. Routes to S-nitroso-hemoglobin formation with heme redox and preferential reactivity in the β subunits. Proc. Natl Acad. Sci. USA 100, 461–466 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Foster, M. W. & Stamler, J. S. New insights into protein S-nitrosylation: mitochondria as a model system. J. Biol. Chem. 279, 25891–25897 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Mulsch, A., Mordvintcev, P. I., Vanin, A. F. & Busse, R. Formation and release of dinitrosyl iron complexes by endothelial cells. Biochem. Biophys. Res. Commun. 196, 1303–1308 (1993).

    Article  CAS  PubMed  Google Scholar 

  32. Vanin, A. F., Mordvintcev, P. I., Hauschildt, S. & Mulsch, A. The relationship between l-arginine-dependent nitric oxide synthesis, nitrite release and dinitrosyl–iron complex formation by activated macrophages. Biochim. Biophys. Acta 1177, 37–42 (1993).

    Article  CAS  PubMed  Google Scholar 

  33. Pawloski, J. R., Hess, D. T. & Stamler, J. S. Export by red blood cells of nitric oxide bioactivity. Nature 409, 622–626 (2001).

    Article  CAS  PubMed  Google Scholar 

  34. Patel, J. M., Zhang, J. & Block, E. R. Nitric oxide-induced inhibition of lung endothelial cell nitric oxide synthase via interaction with allosteric thiols: role of thioredoxin in regulation of catalytic activity. Am. J. Respir. Cell Mol. Biol. 15, 410–419 (1996).

    Article  CAS  PubMed  Google Scholar 

  35. Ravi, K., Brennan, L. A., Levic, S., Ross, P. A. & Black, S. M. S-nitrosylation of endothelial nitric oxide synthase is associated with monomerization and decreased enzyme activity. Proc. Natl Acad. Sci. USA 101, 2619–2624 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kahlos, K., Zhang, J., Block, E. R. & Patel, J. M. Thioredoxin restores nitric oxide-induced inhibition of protein kinase C activity in lung endothelial cells. Mol. Cell. Biochem. 254, 47–54 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Nikitovic, D. & Holmgren, A. S-nitrosoglutathione is cleaved by the thioredoxin system with liberation of glutathione and redox regulating nitric oxide. J. Biol. Chem. 271, 19180–19185 (1996).

    Article  CAS  PubMed  Google Scholar 

  38. Gaston, B. et al. Endogenous nitrogen oxides and bronchodilator S-nitrosothiols in human airways. Proc. Natl Acad. Sci. USA 90, 10957–10961 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Liu, L. et al. A metabolic enzyme for S-nitrosothiol conserved from bacteria to humans. Nature 410, 490–494 (2001).

    Article  CAS  PubMed  Google Scholar 

  40. Sun, J., Xu, L., Eu, J. P., Stamler, J. S. & Meissner, G. Nitric oxide, NOC-12, and S-nitrosoglutathione modulate the skeletal muscle calcium release channel/ryanodine receptor by different mechanisms. An allosteric function for O2 in S-nitrosylation of the channel. J. Biol. Chem. 278, 8184–8189 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. Hess, D. T., Matsumoto, A., Nudelman, R. & Stamler, J. S. S-nitrosylation: spectrum and specificity. Nature Cell Biol. 3, E46–E49 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Campbell, D. L., Stamler, J. S. & Strauss, H. C. Redox modulation of L-type calcium channels in ferret ventricular myocytes. Dual mechanism regulation by nitric oxide and S-nitrosothiols. J. Gen. Physiol. 108, 277–293 (1996).

    Article  CAS  PubMed  Google Scholar 

  43. Matsumoto, A., Comatas, K. E., Liu, L. & Stamler, J. S. Screening for nitric oxide-dependent protein–protein interactions. Science 301, 657–661 (2003). Used yeast two-hybrid screening to show S -nitrosylation-facilitated protein–protein interactions, including interactions of NOS with substrates for S -nitrosylation, which were verified in mammalian cells.

    Article  CAS  PubMed  Google Scholar 

  44. Matsushita, K. et al. Nitric oxide regulates exocytosis by S-nitrosylation of N-ethylmaleimide-sensitive factor. Cell 115, 139–150 (2003). Showed that the activity of NSF, an essential component of most or all membrane trafficking, is regulated in situ by endogenous S -nitrosylation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Stamler, J. S. et al. Blood flow regulation by S-nitrosohemoglobin in the physiological oxygen gradient. Science 276, 2034–2037 (1997).

    Article  CAS  PubMed  Google Scholar 

  46. Eu, J. P., Sun, J., Xu, L., Stamler, J. S. & Meissner, G. The skeletal muscle calcium release channel: coupled O2 sensor and NO signaling functions. Cell 102, 499–509 (2000). Showed that Ca2+ flux through the ryanodine receptor/Ca2+-release channel of skeletal muscle (RyR1) was activated in situ by S -nitrosylation of a single regulatory Cys, and that S -nitrosylation was gated by oxygen-dependent alteration in the redox status of a small additional set of Cys residues.

    Article  CAS  PubMed  Google Scholar 

  47. Lai, T. S. et al. Calcium regulates S-nitrosylation, denitrosylation, and activity of tissue transglutaminase. Biochemistry 40, 4904–4910 (2001).

    Article  CAS  PubMed  Google Scholar 

  48. Jia, L., Bonaventura, C., Bonaventura, J. & Stamler, J. S. S-nitrosohaemoglobin: a dynamic activity of blood involved in vascular control. Nature 380, 221–226 (1996). Showed that haemoglobin is S -nitrosylated endogenously (rather than eliminating nitric oxide) and thereby functions as a source of vasodilatory activity conveyed by red blood cells (which subserves hypoxic vasodilation).

    Article  CAS  PubMed  Google Scholar 

  49. James, P. E., Lang, D., Tufnell-Barret, T., Milsom, A. B. & Frenneaux, M. P. Vasorelaxation by red blood cells and impairment in diabetes. Reduced nitric oxide and oxygen delivery by glycated hemoglobin. Circ. Res. 94, 976–983 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Funai, E. F., Davidson, A., Seligman, S. P. & Finlay, T. H. S-nitrosohemoglobin in the fetal circulation may represent a cycle for blood pressure regulation. Biochem. Biophys. Res. Commun. 239, 875–877 (1997).

    Article  CAS  PubMed  Google Scholar 

  51. Singel, D. J. & Stamler, J. S. Chemical physiology of blood flow regulation by red blood cells: role of nitric oxide and S-nitrosohemoglobin. Annu. Rev. Physiol. Oct 19 2004 (doi:10.1146/annurev.physiol.67.060603.090918).

  52. Stamler, J. S., Toone, E. J., Lipton, S. A. & Sucher, N. J. (S)NO signals: translocation, regulation, and a consensus motif. Neuron 18, 691–696 (1997).

    Article  CAS  PubMed  Google Scholar 

  53. Britto, P. J., Knipling, L. & Wolff, J. The local electrostatic environment determines cysteine reactivity of tubulin. J. Biol. Chem. 277, 29018–29027 (2002).

    Article  CAS  PubMed  Google Scholar 

  54. Bizzozero, O. A., Bixler, H. A. & Pastuszyn, A. Structural determinants influencing the reaction of cysteine-containing peptides with palmitoyl-coenzyme A and other thioesters. Biochim. Biophys. Acta 1545, 278–288 (2001).

    Article  CAS  PubMed  Google Scholar 

  55. Atkins, W. M., Wang, R. W., Bird, A. W., Newton, D. J. & Lu, A. Y. The catalytic mechanism of glutathione S-transferase (GST). Spectroscopic determination of the pKa of Tyr-9 in rat α1-1 GST. J. Biol. Chem. 268, 19188–19191 (1993).

    Article  CAS  PubMed  Google Scholar 

  56. Pérez-Mato, I., Castro, C., Ruiz, F. A., Corrales, F. J. & Mato, J. M. Methionine adenosyltransferase S-nitrosylation is regulated by the basic and acidic amino acids surrounding the target thiol. J. Biol. Chem. 274, 17075–17079 (1999). Showed with site-specific mutation the important role of acidic and basic side chains, proximate to Cys thiol, in targeting S -nitrosylation within protein substrates (transnitrosylation by GSNO).

    Article  PubMed  Google Scholar 

  57. Brenman, J. E. et al. Interaction of nitric oxide synthase with the postsynaptic density protein PSD-95 and α1-syntrophin mediated by PDZ domains. Cell 84, 757–767 (1996).

    Article  CAS  PubMed  Google Scholar 

  58. Kornau, H. C., Schenker, L. T., Kennedy, M. B. & Seeburg, P. H. Domain interaction between NMDA receptor subunits and the postsynaptic density protein PSD-95. Science 269, 1737–1740 (1995).

    Article  CAS  PubMed  Google Scholar 

  59. Lipton, S. A. et al. Cysteine regulation of protein function — as exemplified by NMDA-receptor modulation. Trends Neurosci. 25, 474–480 (2002).

    Article  CAS  PubMed  Google Scholar 

  60. Fang, M. et al. Dexras1: a G protein specifically coupled to neuronal nitric oxide synthase via CAPON. Neuron 28, 183–193 (2000).

    Article  CAS  PubMed  Google Scholar 

  61. Rizzo, M. A. & Piston, D. W. Regulation of β cell glucokinase by S-nitrosylation and association with nitric oxide synthase. J. Cell Biol. 161, 243–248 (2003). Showed that glucokinase is bound to nNOS in pancreatic β-cells, and that physiological activation of nNOS by insulin results in S -nitrosylation of glucokinase, its release from nNOS and activation of kinase activity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Giles, N. M., Giles, G. I. & Jacob, C. Multiple roles of cysteine in biocatalysis. Biochem. Biophys. Res. Commun. 300, 1–4 (2003).

    Article  CAS  PubMed  Google Scholar 

  63. Stamler, J. S., Singel, D. J. & Loscalzo, J. Biochemistry of nitric oxide and its redox-activated forms. Science 258, 1898–1902 (1992).

    Article  CAS  PubMed  Google Scholar 

  64. Mannick, J. B. et al. S-Nitrosylation of mitochondrial caspases. J. Cell Biol. 154, 1111–1116 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Kim, J. E. & Tannenbaum, S. R. S-nitrosation regulates the activation of endogenous procaspase-9 in HT-29 human colon carcinoma cells. J. Biol. Chem. 279, 9758–9764 (2004).

    Article  CAS  PubMed  Google Scholar 

  66. Caselli, A., Chiarugi, P., Camici, G., Manao, G. & Ramponi, G. In vivo inactivation of phosphotyrosine protein phosphatases by nitric oxide. FEBS Lett. 374, 249–252 (1995).

    Article  CAS  PubMed  Google Scholar 

  67. Callsen, D., Sandau, K. B. & Brune, B. Nitric oxide and superoxide inhibit platelet-derived growth factor receptor phosphotyrosine phosphatases. Free Radic. Biol. Med. 26, 1544–1553 (1999).

    Article  CAS  PubMed  Google Scholar 

  68. Xian, M. et al. Inhibition of protein tyrosine phosphatases by low-molecular-weight S-nitrosothiols and S-nitrosylated human serum albumin. Biochem. Biophys. Res. Commun. 268, 310–314 (2000).

    Article  CAS  PubMed  Google Scholar 

  69. Li, S. & Whorton, A. R. Regulation of protein tyrosine phosphatase 1B in intact cells by S-nitrosothiols. Arch. Biochem. Biophys. 410, 269–279 (2003).

    Article  CAS  PubMed  Google Scholar 

  70. Mikkelsen, R. B. & Wardman, P. Biological chemistry of reactive oxygen and nitrogen and radiation-induced signal transduction mechanisms. Oncogene 22, 5734–5754 (2003).

    Article  CAS  PubMed  Google Scholar 

  71. Leiper, J., Murray-Rust, J., McDonald, N. & Vallance, P. S-nitrosylation of dimethylarginine dimethylaminohydrolase regulates enzyme activity: further interactions between nitric oxide synthase and dimethylarginine dimethylaminohydrolase. Proc. Natl Acad. Sci. USA 99, 13527–13532 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Hao, G., Xie, L. & Gross, S. S. Argininosuccinate synthetase is reversibly inactivated by S-nitrosylation in vitro and in vivo. J. Biol. Chem. 279, 36192–36200 (2004).

    Article  CAS  PubMed  Google Scholar 

  73. Bauer, P. M., Buga, G. M., Fukuto, J. M., Pegg, A. E. & Ignarro, L. J. Nitric oxide inhibits ornithine decarboxylase via S-nitrosylation of cysteine 360 in the active site of the enzyme. J. Biol. Chem. 276, 34458–34464 (2001).

    Article  CAS  PubMed  Google Scholar 

  74. Hillary, R. A. & Pegg, A. E. Decarboxylases involved in polyamine biosynthesis and their inactivation by nitric oxide. Biochim. Biophys. Acta 1647, 161–166 (2003).

    Article  CAS  PubMed  Google Scholar 

  75. Haendeler, J. et al. Redox regulatory and anti-apoptotic functions of thioredoxin depend on S-nitrosylation at cysteine 69. Nature Cell Biol. 4, 743–749 (2002).

    Article  CAS  PubMed  Google Scholar 

  76. Sumbayev, V. V. S-nitrosylation of thioredoxin mediates activation of apoptosis signal-regulating kinase 1. Arch. Biochem. Biophys. 415, 133–136 (2003).

    Article  CAS  PubMed  Google Scholar 

  77. Park, H. S. et al. Inhibition of apoptosis signal-regulating kinase 1 (ASK1) by nitric oxide through a thiol-redox mechanism. J. Biol. Chem. 279, 7584–7590 (2003).

    Article  PubMed  CAS  Google Scholar 

  78. Park, H. S., Huh, S. H., Kim, M. S., Lee, S. H. & Choi, E. J. Nitric oxide negatively regulates c-Jun N-terminal kinase/stress-activated protein kinase by means of S-nitrosylation. Proc. Natl Acad. Sci. USA 97, 14382–14387 (2000). Showed in intact cells that endogenously produced NO can suppress activity of a protein kinase, JNK, which could be ascribed to S -nitrosylation of a single regulatory cysteine.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Lander, H. M., Ogiste, J. S., Pearce, S. F., Levi, R. & Novogrodsky, A. Nitric oxide-stimulated guanine nucleotide exchange on p21ras. J. Biol. Chem. 270, 7017–7020 (1995).

    Article  CAS  PubMed  Google Scholar 

  80. dela Torre, A., Schroeder, R. A., Bartlett, S. T. & Kuo, P. C. Differential effects of nitric oxide-mediated S-nitrosylation on p50 and c-jun DNA binding. Surgery 124, 137–141 (1998).

    Article  CAS  Google Scholar 

  81. Nikitovic, D., Holmgren, A. & Spyrou, G. Inhibition of AP-1 DNA binding by nitric oxide involving conserved cysteine residues in Jun and Fos. Biochem. Biophys. Res. Commun. 242, 109–112 (1998).

    Article  CAS  PubMed  Google Scholar 

  82. Monteiro, H. P., Gruia-Gray, J., Peranovich, T. M., de Oliveira, L. C. & Stern, A. Nitric oxide stimulates tyrosine phosphorylation of focal adhesion kinase, Src kinase, and mitogen-activated protein kinases in murine fibroblasts. Free Radic. Biol. Med. 28, 174–182 (2000).

    Article  CAS  PubMed  Google Scholar 

  83. Akhand, A. A. et al. Nitric oxide controls src kinase activity through a sulfhydryl group modification-mediated Tyr-527-independent and Tyr-416-linked mechanism. J. Biol. Chem. 274, 25821–25826 (1999).

    Article  CAS  PubMed  Google Scholar 

  84. Yun, H. Y., Gonzalez-Zulueta, M., Dawson, V. L. & Dawson, T. M. Nitric oxide mediates N-methyl-D-aspartate receptor-induced activation of p21ras. Proc. Natl Acad. Sci. USA 95, 5773–5778 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Lin, Y. F., Raab-Graham, K., Jan, Y. N. & Jan, L. Y. NO stimulation of ATP-sensitive potassium channels: involvement of Ras/mitogen-activated protein kinase pathway and contribution to neuroprotection. Proc. Natl Acad. Sci. USA 101, 7799–7804 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Jaffrey, S. R., Fang, M. & Snyder, S. H. Nitrosopeptide mapping: a novel methodology reveals S-nitrosylation of dexras1 on a single cysteine residue. Chem. Biol. 9, 1329–1335 (2002).

    Article  CAS  PubMed  Google Scholar 

  87. Estrada, C. et al. Nitric oxide reversibly inhibits the epidermal growth factor receptor tyrosine kinase. Biochem. J. 326, 369–376 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Nozik-Grayck, E. et al. Pulmonary vasoconstriction by serotonin is inhibited by S-nitrosoglutathione. Am. J. Physiol. Lung Cell. Mol. Physiol. 282, L1057–L1065 (2002).

    Article  CAS  PubMed  Google Scholar 

  89. Lipton, S. A. et al. A redox-based mechanism for the neuroprotective and neurodestructive effects of nitric oxide and related nitroso-compounds. Nature 364, 626–632 (1993).

    Article  CAS  PubMed  Google Scholar 

  90. Choi, Y. B. et al. Molecular basis of NMDA receptor-coupled ion channel modulation by S-nitrosylation. Nature Neurosci. 3, 15–21 (2000).

    Article  CAS  PubMed  Google Scholar 

  91. Sun, J., Xin, C., Eu, J. P., Stamler, J. S. & Meissner, G. Cysteine-3635 is responsible for skeletal muscle ryanodine receptor modulation by NO. Proc. Natl Acad. Sci. USA 98, 11158–11162 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Eu, J. P. et al. Concerted regulation of skeletal muscle contractility by oxygen tension and endogenous nitric oxide. Proc. Natl Acad. Sci. USA 100, 15229–15234 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Aracena, P., Sanchez, G., Donoso, P., Hamilton, S. L. & Hidalgo, C. S-glutathionylation decreases Mg2+ inhibition and S-nitrosylation enhances Ca2+ activation of RyR1 channels. J. Biol. Chem. 278, 42927–42935 (2003).

    Article  CAS  PubMed  Google Scholar 

  94. Sun, J., Xu, L., Eu, J. P., Stamler, J. S. & Meissner, G. Classes of thiols that influence the activity of the skeletal muscle calcium release channel. J. Biol. Chem. 276, 15625–15630 (2001).

    Article  CAS  PubMed  Google Scholar 

  95. Zable, A. C., Favero, T. G. & Abramson, J. J. Glutathione modulates ryanodine receptor from skeletal muscle sarcoplasmic reticulum. Evidence for redox regulation of the Ca2+ release mechanism. J. Biol. Chem. 272, 7069–7077 (1997).

    Article  CAS  PubMed  Google Scholar 

  96. Xu, L., Eu, J. P., Meissner, G. & Stamler, J. S. Activation of the cardiac calcium release channel (ryanodine receptor) by poly-S-nitrosylation. Science 279, 234–237 (1998).

    Article  CAS  PubMed  Google Scholar 

  97. Xu, K. Y., Huso, D. L., Dawson, T. M., Bredt, D. S. & Becker, L. C. Nitric oxide synthase in cardiac sarcoplasmic reticulum. Proc. Natl Acad. Sci. USA 96, 657–662 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Broillet, M. C. & Firestein, S. Direct activation of the olfactory cyclic nucleotide-gated channel through modification of sulfhydryl groups by NO compounds. Neuron 16, 377–385 (1996).

    Article  CAS  PubMed  Google Scholar 

  99. Broillet, M. C. A single intracellular cysteine residue is responsible for the activation of the olfactory cyclic nucleotide-gated channel by NO. J. Biol. Chem. 275, 15135–15141 (2000).

    Article  CAS  PubMed  Google Scholar 

  100. Gao, C. et al. S-nitrosylation of heterogeneous nuclear ribonucleoprotein A/B regulates osteopontin transcription in endotoxin-stimulated murine macrophages. J. Biol. Chem. 279, 11236–11243 (2004).

    Article  CAS  PubMed  Google Scholar 

  101. Melillo, G. et al. A hypoxia-responsive element mediates a novel pathway of activation of the inducible nitric oxide synthase promoter. J. Exp. Med. 182, 1683–1693 (1995).

    Article  CAS  PubMed  Google Scholar 

  102. Palmer, L. A., Semenza, G. L., Stoler, M. H. & Johns, R. A. Hypoxia induces type II NOS gene expression in pulmonary artery endothelial cells via HIF-1. Am. J. Physiol. 274, L212–L219 (1998).

    CAS  PubMed  Google Scholar 

  103. Ambs, S., Hussain, S. P. & Harris, C. C. Interactive effects of nitric oxide and the p53 tumor suppressor gene in carcinogenesis and tumor progression. FASEB J. 11, 443–448 (1997).

    Article  CAS  PubMed  Google Scholar 

  104. Davis, M. E., Grumbach, I. M., Fukai, T., Cutchins, A. & Harrison, D. G. Shear stress regulates endothelial nitric-oxide synthase promoter activity through nuclear factor κB binding. J. Biol. Chem. 279, 163–168 (2004).

    Article  CAS  PubMed  Google Scholar 

  105. Lowenstein, C. J. et al. Macrophage nitric oxide synthase gene: two upstream regions mediate induction by interferon γ and lipopolysaccharide. Proc. Natl Acad. Sci. USA 90, 9730–9734 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Xie, Q. W., Kashiwabara, Y. & Nathan, C. Role of transcription factor NF-κB/Rel in induction of nitric oxide synthase. J. Biol. Chem. 269, 4705–4708 (1994).

    Article  CAS  PubMed  Google Scholar 

  107. Hausladen, A., Privalle, C. T., Keng, T., DeAngelo, J. & Stamler, J. S. Nitrosative stress: activation of the transcription factor OxyR. Cell 86, 719–729 (1996).

    Article  CAS  PubMed  Google Scholar 

  108. Kullik, I., Toledano, M. B., Tartaglia, L. A. & Storz, G. Mutational analysis of the redox-sensitive transcriptional regulator OxyR: regions important for oxidation and transcriptional activation. J. Bacteriol. 177. 1275–1284 (1995)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Kim, S. O. et al. OxyR: a molecular code for redox-related signaling. Cell 109, 383–396 (2002). Showed that a single regulatory Cys within the bacterial transcription factor OxyR is subject not only to S -nitrosylation but also to additional NO-dependent and NO-independent oxidative modifications, with differential effects on DNA binding and transcriptional response.

    Article  CAS  PubMed  Google Scholar 

  110. Pugh, C. W. & Ratcliffe, P. J. Regulation of angiogenesis by hypoxia: role of the HIF system. Nature Med. 9, 677–684 (2003).

    Article  CAS  PubMed  Google Scholar 

  111. Sumbayev, V. V., Budde, A., Zhou, J. & Brune, B. HIF- 1α protein as a target for S-nitrosation. FEBS Lett. 535, 106–112 (2003).

    Article  CAS  PubMed  Google Scholar 

  112. Yasinska, I. M. & Sumbayev, V. V. S-nitrosation of Cys-800 of HIF-1α protein activates its interaction with p300 and stimulates its transcriptional activity. FEBS Lett. 549, 105–109 (2003).

    Article  CAS  PubMed  Google Scholar 

  113. Palmer, L. A., Gaston, B. & Johns, R. A. Normoxic stabilization of hypoxia-inducible factor-1 expression and activity: redox-dependent effect of nitrogen oxides. Mol. Pharmacol. 58, 1197–1203 (2000). Provided an initial demonstration, with HIF1, that NO could control the activity of transcription factors by regulating their stabiltity (ubiquitylation and proteasomal degradation).

    Article  CAS  PubMed  Google Scholar 

  114. Sandau, K. B., Fandrey, J. & Brune, B. Accumulation of HIF-1α under the influence of nitric oxide. Blood 97, 1009–1015 (2001).

    Article  CAS  PubMed  Google Scholar 

  115. Metzen, E., Zhou, J., Jelkmann, W., Fandrey, J. & Brune, B. Nitric oxide impairs normoxic degradation of HIF-1α by inhibition of prolyl hydroxylases. Mol. Biol. Cell 14, 3470–3481 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Yao, D. et al. Nitrosative stress linked to sporadic Parkinson's disease: S-nitrosylation of parkin regulates its E3 ubiquitin ligase activity. Proc. Natl Acad. Sci. USA 101, 10810–10814 (2004). Showed, together with reference 117, that the activity of the neuronal E3 ubiquitin ligase parkin is regulated by S -nitrosylation in situ as a result of nitrosative stress induced experimentally or occuring endogenously in the brains of Parkinson's disease patients.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Chung, K. K. K. et al. S-nitrosylation of parkin regulates ubiquitination and compromises parkin's protective function. Science 304, 1328–1331 (2004).

    Article  CAS  PubMed  Google Scholar 

  118. Michael, D. & Oren, M. The p53–Mdm2 module and the ubiquitin system. Semin. Cancer Biol. 13, 49–58 (2003).

    Article  CAS  PubMed  Google Scholar 

  119. Calmels, S., Hainaut, P. & Ohshima, H. Nitric oxide induces conformational and functional modifications of wild-type p53 tumor suppressor protein. Cancer Res. 57, 3365–3369 (1997).

    CAS  PubMed  Google Scholar 

  120. Brune, B., von Knethen, A. & Sandau, K. B. Transcription factors p53 and HIF-1α as targets of nitric oxide. Cell. Signal. 13, 525–533 (2001).

    Article  CAS  PubMed  Google Scholar 

  121. Schonhoff, C. M., Daou, M. C., Jones, S. N., Schiffer, C. A. & Ross, A. H. Nitric oxide-mediated inhibition of Hdm2–p53 binding. Biochemistry 41, 13570–13574 (2002).

    Article  CAS  PubMed  Google Scholar 

  122. Karin, M. & Ben-Neriah, Y. Phosphorylation meets ubiquitination: the control of NFκB activity. Annu. Rev. Immunol. 18, 621–663 (2000).

    Article  CAS  PubMed  Google Scholar 

  123. dela Torre, A., Schroeder, R. A., Punzalan, C. & Kuo, P. C. Endotoxin-mediated S-nitrosylation of p50 alters NF-κB-dependent gene transcription in ANA-1 murine macrophages. J. Immunol. 162, 4101–4108 (1999).

    CAS  Google Scholar 

  124. Park, S. K., Lin, H. L. & Murphy, S. Nitric oxide regulates nitric oxide synthase-2 gene expression by inhibiting NF-κB binding to DNA. Biochem. J. 322, 609–613 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. dela Torre, A., Schroeder, R. A. & Kuo, P. C. Alteration of NF-κB p50 DNA binding kinetics by S-nitrosylation. Biochem. Biophys. Res. Commun. 238, 703–706 (1997).

    Article  CAS  Google Scholar 

  126. Matthews, J. R., Botting, C. H., Panico, M., Morris, H. R. & Hay, R. T. Inhibition of NF-κB DNA binding by nitric oxide. Nucleic Acids Res. 24, 2236–2242 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Marshall, H. E. & Stamler, J. S. Inhibition of NF-κB by S-nitrosylation. Biochemistry 40, 1688–1693 (2001).

    Article  CAS  PubMed  Google Scholar 

  128. Peng, H. B., Libby, P. & Liao, J. K. Induction and stabilization of IκBα by nitric oxide mediates inhibition of NF-κB. J. Biol. Chem. 270, 14214–14219 (1995).

    Article  CAS  PubMed  Google Scholar 

  129. Marshall, H. E. & Stamler, J. S. Nitrosative stress-induced apoptosis through inhibition of NF-κB. J. Biol. Chem. 277, 34223–34228 (2002).

    Article  CAS  PubMed  Google Scholar 

  130. Reynaert, N. L. et al. Nitric oxide represses inhibitory κB kinase through S-nitrosylation. Proc. Natl Acad. Sci. USA 101, 8945–8950 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Pantopoulos, K. & Hentze, M. W. Nitric oxide signaling to iron-regulatory protein: direct control of ferritin mRNA translation and transferrin receptor mRNA stability in transfected fibroblasts. Proc. Natl Acad. Sci. USA 92, 1267–1271 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Kim, S. & Ponka, P. Effects of interferon-γ and lipopolysaccharide on macrophage iron metabolism are mediated by nitric oxide-induced degradation of iron regulatory protein 2. J. Biol. Chem. 275, 6220–6226 (2000).

    Article  CAS  PubMed  Google Scholar 

  133. Bouton, C., Oliveira, L. & Drapier, J. C. Converse modulation of IRP1 and IRP2 by immunological stimuli in murine RAW 264.7 macrophages. J. Biol. Chem. 273, 9403–9408 (1998).

    Article  CAS  PubMed  Google Scholar 

  134. Kim, S., Wing, S. S. & Ponka, P. S-nitrosylation of IRP2 regulates its stability via the ubiquitin-proteasome pathway. Mol. Cell. Biol. 24, 330–337 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. LaVaute, T. et al. Targeted deletion of the gene encoding iron regulatory protein-2 causes misregulation of iron metabolism and neurodegenerative disease in mice. Nature Genet. 27, 209–214 (2001).

    Article  CAS  PubMed  Google Scholar 

  136. Jaffrey, S. R., Cohen, N. A., Rouault, T. A., Klausner, R. D. & Snyder, S. H. The iron-responsive element binding protein: a target for synaptic actions of nitric oxide. Proc. Natl Acad. Sci. USA 91, 12994–12998 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Dudev, T. & Lim, C. Factors governing the protonation state of cysteines in proteins: an Ab initio/CDM study. J. Am. Chem. Soc. 124, 6759–6766 (2002).

    Article  CAS  PubMed  Google Scholar 

  138. Kroncke, K. D., Klotz, L. O., Suschek, C. V. & Sies, H. Comparing nitrosative versus oxidative stress toward zinc finger-dependent transcription. Unique role for NO. J. Biol. Chem. 277, 13294–13301 (2002).

    Article  CAS  PubMed  Google Scholar 

  139. Pearce, L. L. et al. Role of metallothionein in nitric oxide signaling as revealed by a green fluorescent fusion protein. Proc. Natl Acad. Sci. USA 97, 477–482 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Bossy-Wetzel, E. et al. Crosstalk between nitric oxide and zinc pathways to neuronal cell death involving mitochondrial dysfunction and p38-activated K+ channels. Neuron 41, 351–365 (2004).

    Article  CAS  PubMed  Google Scholar 

  141. Gu, Z. et al. S-nitrosylation of matrix metalloproteinases: signaling pathway to neuronal cell death. Science 297, 1186–1190 (2002).

    Article  CAS  PubMed  Google Scholar 

  142. Zhang, Z. et al. Activation of tumor necrosis factor-α-converting enzyme-mediated ectodomain shedding by nitric oxide. J. Biol. Chem. 275, 15839–15844 (2000).

    Article  CAS  PubMed  Google Scholar 

  143. Datta, B. et al. Red blood cell nitric oxide as an endocrine vasoregulator: a potential role in congestive heart failure. Circulation 109, 1339–1342 (2004).

    Article  CAS  PubMed  Google Scholar 

  144. Massy, Z. A. et al. Increased plasma S-nitrosothiol concentrations predict cardiovascular outcomes among patients with end-stage renal disease: a prospective study. J. Am. Soc. Nephrol. 15, 470–476 (2004).

    Article  CAS  PubMed  Google Scholar 

  145. Gow, A. J., Buerk, D. G. & Ischiropoulos, H. A novel reaction mechanism for the formation of S-nitrosothiol in vivo. J. Biol. Chem. 272, 2841–2845 (1997).

    Article  CAS  PubMed  Google Scholar 

  146. Houk, K. N. et al. Nitroxyl disulfides, novel intermediates in transnitrosation reactions. J. Am. Chem. Soc. 125, 6972–6976 (2003).

    Article  CAS  PubMed  Google Scholar 

  147. Bartberger, M. D. et al. Theory, spectroscopy, and crystallographic analysis of S-nitrosothiols: conformational distribution dictates spectroscopic behavior. J. Am. Chem. Soc. 122, 5889–5890 (2000).

    Article  CAS  Google Scholar 

  148. Chan, N. L., Kavanaugh, J. S., Rogers, P. H. & Arnone, A. Crystallographic analysis of the interaction of nitric oxide with quaternary-T human hemoglobin. Biochemistry 43, 118–132 (2004).

    Article  CAS  PubMed  Google Scholar 

  149. Liu, X., Miller, M. J., Joshi, M. S., Thomas, D. D. & Lancaster, J. R. Jr. Accelerated reaction of nitric oxide with O2 within the hydrophobic interior of biological membranes. Proc. Natl Acad. Sci. USA 95, 2175–2179 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Nedospasov, A., Rafikov, R., Beda, N. & Nudler, E. An autocatalytic mechanism of protein nitrosylation. Proc. Natl Acad. Sci. USA 97, 13543–13548 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Jourd'heuil, D., Jourd'heuil, F. L. & Feelisch, M. Oxidation and nitrosation of thiols at low micromolar exposure to nitric oxide. Evidence for a free radical mechanism. J. Biol. Chem. 278, 15720–15726 (2003).

    Article  CAS  PubMed  Google Scholar 

  152. Liu, L. et al. Inactivation of annexin II tetramer by S-nitrosoglutathione. Eur. J. Biochem. 269, 4277–4286 (2002).

    Article  CAS  PubMed  Google Scholar 

  153. Ji, Y., Toader, V. & Bennett, B. M. Regulation of microsomal and cytosolic glutathione S-transferase activities by S-nitrosylation. Biochem. Pharmacol. 63, 1397–1404 (2002).

    Article  CAS  PubMed  Google Scholar 

  154. Ventura, A. & Pelicci, P. G. Semaphorins: green light for redox signaling? Sci. STKE 2002, PE44 (2002).

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jonathan S. Stamler.

Ethics declarations

Competing interests

Dr Stamler and Dr Matsumoto are inventors on patents that concern NO biology, and Dr Stamler is involved in development of NO-based technologies.

Related links

Related links

DATABASES

Swiss-Prot

ASK1

CAPON

caspase-3

caspase-8

caspase-9

Dexras1

eNOS

HDM2

HIF1α

HIF2α

IKKα

IKKβ

IKKγ

iNOS

IRP1

IRP2

JNK1

JNK3

MKK3

MKK6

MMP9

nNOS

OxyR

p21Ras

p53

p60Src

PSD95

RyR1

RyR2

SOD

TACE

TNFα

TRX

FURTHER INFORMATION

eMOTIF

Glossary

NO SYNTHASE

(NOS). Mammals have three NO synthases that generate NO from Arg — NOS1 or nNOS, NOS2 or iNOS and NOS3 or eNOS — one or more of which reside, or can be induced, in most or all cell types. NOS homologues are distributed broadly across the phylogeny.

S-NITROSO-GLUTATHIONE

(GSNO). The main non-protein S-nitrosothiol (SNO) in cells, which functions in an equilibrium with protein SNOs (the equilibrium is determined in part by the enzyme GSNO-reductase, which metabolizes GSNO (see Table 1)).

OXIDOREDUCTASE

An enzyme that catalyses oxidation–reduction reactions, which entail the transfer of electrons from a substrate that becomes oxidized (electron donor) to a substrate that becomes reduced (electron acceptor).

NITROSATIVE AND OXIDATIVE STRESS

The dysregulated production and/or metabolism of reactive nitrogen and/or oxygen species, which generate nitrosative and/or oxidative chemistries that can result in disrupted cellular signalling, injury and death.

NUCLEOPHILICITY

Nucleophilic groups such as the sulphur group of Cys are electron rich, and the degree of nucleophilicity is a measure of their reactivity towards electrophiles (electron-deficient species such as NO+), as exemplified by the increased reactivity of cysteine thiolate versus sulfhydryl.

ALLOSTERIC REGULATOR

An ion or small molecule that reacts with and thereby modulates the conformation and function of proteins. A number of such allosteric regulators, including O2 and Ca2+, have been implicated in the regulation of protein S-nitrosylation.

METALLOPROTEIN

A protein that contains a metal ion (or ions; such as Fe2+, Cu2+ or Zn2+) as a prosthetic group, which is coordinated by amino-acid side chains.

TAUTOMERIC

Describes the relationship between two structural isomers of a molecule that are in chemical equilibrium, which includes the two forms that result from the intramolecular transfer of an acidic proton.

REACTIVE OXYGEN SPECIES

Reduced derivatives of molecular oxygen (O2), including, in particular, the superoxide radical (O2•−) and hydrogen peroxide (H2O2), which can have significant reactivity towards biological macromolecules and towards other reactive small molecules.

POLYAMINE

A metabolic product of Arg (the substrate for NO synthases), which is generated through a highly regulated sequence of enzymatic reactions.

WEIBEL–PALADE BODY

A secretory vesicle containing agents, including the von Willebrand factor, which are exocytosed from endothelial cells following inflammatory stimulation.

SARCOPLASMIC RETICULUM (SR).

A specialized form of endoplasmic reticulum in muscle cells that sequesters and releases Ca2+ to control muscle contractility.

GSH/GSSG

The ratio of reduced to oxidized (disulphide-linked) glutathione. This ratio is a principal indicator of the redox state of a cell or subcellular compartment.

INOTROPIC

Influencing muscle contractility.

NORMOXIA

Ambient O2 (21%) or, in reference to tissue, O2 concentrations that represent the normal physiological state (2–3%).

LEWIS ACID

An electron-pair acceptor that can participate as one member of a conjugate acid–base pair in acid–base reactions.

METALLOTHIONEIN

A small Cys-rich protein that binds metal ions — in particular, Zn2+ and/or Cu2+ — at least in part, through complex formation with cysteine thiolate.

EXTRACELLULAR MATRIX METALLOPROTEINASE

(MMP). A Zn2+-dependent proteolytic enzyme that has an extracellular active site, which is capable of breaking down extracellular-matrix components.

METALLOPROTEINASE/DISINTEGRIN (ADAM) FAMILY

A family of extracellular metalloproteinases that are implicated in the proteolytic processing of membrane-bound substrates, which results in ectodomain shedding, and are named after their characteristic ADAM-domain structure.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hess, D., Matsumoto, A., Kim, SO. et al. Protein S-nitrosylation: purview and parameters. Nat Rev Mol Cell Biol 6, 150–166 (2005). https://doi.org/10.1038/nrm1569

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm1569

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