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:

Beyond oxidative stress: an immunologist's guide to reactive oxygen species

Key Points

  • Reactive oxygen species (ROS) include superoxide, hydrogen peroxide, singlet oxygen, ozone, hypohalous acids and organic peroxides. They interact with and share some of the actions of other classes of small, reactive, endogenous signalling molecules — reactive nitrogen species such as NO and NO2; H2S or its anion, HS; and carbon monoxide.

  • ROS can both promote and prevent cell death, cancer, ageing and inflammation. For example, ROS mediate inflammasome activation, but patients with chronic granulomatous disease, who lack a functional form of a principal ROS-producing enzyme, NADPH oxidase 2 (NOX2), demonstrate considerable susceptibility to infection, as well as non-resolving inflammation.

  • The numerous enzymatic sources of ROS include mitochondria and multiple isoforms of NOXs. The first NOX, now called NOX2, was discovered in neutrophils, but NOXs contribute to signal transduction in diverse cell types. ROS are produced following B and T cell receptor stimulation and can dictate whether T cell activation is fostered or impeded.

  • Many antioxidant systems contribute to the regulation of ROS, including superoxide dismutases, catalases and the enzymes of the glutathione redox cycle, which reflects the widespread functional effects of ROS.

  • Reactions involving ROS demonstrate atomic rather than molecular specificity. That is, ROS preferentially react with certain types of atoms and most readily with a subset of those atoms, but the atomic targets of ROS are distributed in many different macromolecules. For example, ROS preferentially react with the sulphur atom in some but not other cysteine residues; the cysteine thiols that are most susceptible include many that participate in enzyme active sites, such as in phosphatases. This kind of specificity equips ROS to influence many different signalling pathways simultaneously.

  • The immunosuppressive capacity of myeloid-derived suppressor cells and regulatory T cells results in part from their production of ROS. Tumour cells also produce ROS, which can contribute to their immunosuppressive and metastatic potential.

Abstract

Reactive oxygen species (ROS) react preferentially with certain atoms to modulate functions ranging from cell homeostasis to cell death. Molecular actions include both inhibition and activation of proteins, mutagenesis of DNA and activation of gene transcription. Cellular actions include promotion or suppression of inflammation, immunity and carcinogenesis. ROS help the host to compete against microorganisms and are also involved in intermicrobial competition. ROS chemistry and their pleiotropy make them difficult to localize, to quantify and to manipulate — challenges we must overcome to translate ROS biology into medical advances.

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: The broad range of ROS signalling is influenced by ROS production and catabolism, and by cellular adaptation.
Figure 2: ROS and their atomic specificity.
Figure 3: Examples of transcriptional regulation by ROS acting at the plasma membrane or in the cytosol.
Figure 4: Regulation of HIF1α by mitochondrial ROS production during hypoxia.
Figure 5: Regulation of transcription through DNA targeting by intranuclear ROS.

Similar content being viewed by others

References

  1. Nathan, C. & Ding, A. Snapshot: reactive oxygen intermediates (ROI). Cell 140, 951–951.e2 (2010).

    Article  PubMed  Google Scholar 

  2. Nishida, M. et al. Hydrogen sulfide anion regulates redox signaling via electrophile sulfhydration. Nature Chem. Biol. 8, 714–724 (2012).

    Article  CAS  Google Scholar 

  3. Finkel, T. From sulfenylation to sulfhydration: what a thiolate needs to tolerate. Sci. Signal. 5, pe10 (2012).

    Article  PubMed  CAS  Google Scholar 

  4. Paul, B. D. & Snyder, S. H. H2S signalling through protein sulfhydration and beyond. Nature Rev. Mol. Cell. Biol. 13, 499–507 (2012).

    Article  CAS  Google Scholar 

  5. Wink, D. A. et al. Nitric oxide and redox mechanisms in the immune response. J. Leuk. Biol. 89, 873–891 (2011).

    Article  CAS  Google Scholar 

  6. Steinhubl, S. R. Why have antioxidants failed in clinical trials? Am. J. Cardiol. 101, 14D–19D (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Brennan, M. L. & Hazen, S. L. Emerging role of myeloperoxidase and oxidant stress markers in cardiovascular risk assessment. Curr. Opin. Lipidol. 14, 353–359 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Bae, Y. S., Oh, H., Rhee, S. G. & Yoo, Y. D. Regulation of reactive oxygen species generation in cell signaling. Mol. Cells 32, 491–509 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Finkel, T. Signal transduction by mitochondrial oxidants. J. Biol. Chem. 287, 4434–4440 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Jiang, F., Zhang, Y. & Dusting, G. J. NADPH oxidase-mediated redox signaling: roles in cellular stress response, stress tolerance, and tissue repair. Pharmacol. Rev. 63, 218–242 (2011).

    Article  CAS  PubMed  Google Scholar 

  11. Lambeth, J. D. NOX enzymes and the biology of reactive oxygen. Nature Rev. Immunol. 4, 181–189 (2004).

    Article  CAS  Google Scholar 

  12. Aguirre, J. & Lambeth, J. D. Nox enzymes from fungus to fly to fish and what they tell us about Nox function in mammals. Free Radic. Biol. Med. 49, 1342–1353 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Imlay, J. A. Cellular defenses against superoxide and hydrogen peroxide. Annu. Rev. Biochem. 77, 755–776 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yazdanpanah, B. et al. Riboflavin kinase couples TNF receptor 1 to NADPH oxidase. Nature 460, 1159–1163 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Mailloux, R. J. & Harper, M. E. Uncoupling proteins and the control of mitochondrial reactive oxygen species production. Free Radic. Biol. Med. 51, 1106–1115 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Corzo, C. A. et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J. Immunol. 182, 5693–5701 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Radi, R., Beckman, J. S., Bush, K. M. & Freeman, B. A. Peroxynitrite oxidation of sulfhydryls. The cytotoxic potential of superoxide and nitric oxide. J. Biol. Chem. 266, 4244–4250 (1991).

    CAS  PubMed  Google Scholar 

  18. Gonzalez-Nieto, D. et al. Connexin-43 in the osteogenic BM niche regulates its cellular composition and the bidirectional traffic of hematopoietic stem cells and progenitors. Blood 119, 5144–5154 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Holmgren, A. & Lu, J. Thioredoxin and thioredoxin reductase: current research with special reference to human disease. Biochem. Biophys. Res. Commun. 396, 120–124 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Weissbach, H. et al. Peptide methionine sulfoxide reductase: structure, mechanism of action, and biological function. Arch. Biochem. Biophys. 397, 172–178 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Bryk, R., Griffin, P. & Nathan, C. Peroxynitrite reductase activity of bacterial peroxiredoxins. Nature 407, 211–215 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Morgan, B. et al. Multiple glutathione disulfide removal pathways mediate cytosolic redox homeostasis. Nature Chem. Biol. 9, 119–125 (2012).

    Article  CAS  Google Scholar 

  23. Anastasiou, D. et al. Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses. Science 334, 1278–1283 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. O'Donnell-Tormey, J., Nathan, C. F., Lanks, K., DeBoer, C. J. & de la Harpe, J. Secretion of pyruvate. An antioxidant defense of mammalian cells. J. Exp. Med. 165, 500–514 (1987).

    Article  CAS  PubMed  Google Scholar 

  25. Bertini, R. et al. Thioredoxin, a redox enzyme released in infection and inflammation, is a unique chemoattractant for neutrophils, monocytes, and T cells. J. Exp. Med. 189, 1783–1789 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shichita, T. et al. Peroxiredoxin family proteins are key initiators of post-ischemic inflammation in the brain. Nature Med. 18, 911–917 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Seifert, U. et al. Immunoproteasomes preserve protein homeostasis upon interferon-induced oxidative stress. Cell 142, 613–624 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Scherz-Shouval, R. & Elazar, Z. Regulation of autophagy by ROS: physiology and pathology. Trends Biochem. Sci. 36, 30–38 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Thorpe, G. W., Fong, C. S., Alic, N., Higgins, V. J. & Dawes, I. W. Cells have distinct mechanisms to maintain protection against different reactive oxygen species: oxidative-stress-response genes. Proc. Natl Acad. Sci. USA 101, 6564–6569 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Nathan, C. Specificity of a third kind: reactive oxygen and nitrogen intermediates in cell signaling. J. Clin. Invest. 111, 769–778 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ferrer-Sueta, G. et al. Factors affecting protein thiol reactivity and specificity in peroxide reduction. Chem. Res. Toxicol. 24, 434–450 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. Winterbourn, C. C. & Hampton, M. B. Thiol chemistry and specificity in redox signaling. Free Radic. Biol. Med. 45, 549–561 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Kuiper, J. W., Sun, C., Magalhaes, M. A. & Glogauer, M. Rac regulates PtdInsP3 signaling and the chemotactic compass through a redox-mediated feedback loop. Blood 118, 6164–6171 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Paulsen, C. E. et al. Peroxide-dependent sulfenylation of the EGFR catalytic site enhances kinase activity. Nature Chem. Biol. 8, 57–64 (2012).

    Article  CAS  Google Scholar 

  35. Wani, R. et al. Isoform-specific regulation of Akt by PDGF-induced reactive oxygen species. Proc. Natl Acad. Sci. USA 108, 10550–10555 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Guo, Z., Kozlov, S., Lavin, M. F., Person, M. D. & Paull, T. T. ATM activation by oxidative stress. Science 330, 517–521 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Erickson, J. R. et al. A dynamic pathway for calcium-independent activation of CaMKII by methionine oxidation. Cell 133, 462–474 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Burgoyne, J. R. et al. Cysteine redox sensor in PKGIa enables oxidant-induced activation. Science 317, 1393–1397 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Kroncke, K. D. & Klotz, L. O. Zinc fingers as biologic redox switches? Antioxid. Redox Signal. 11, 1015–1027 (2009).

    Article  PubMed  CAS  Google Scholar 

  40. de Keizer, P. L., Burgering, B. M. & Dansen, T. B. Forkhead box O as a sensor, mediator, and regulator of redox signaling. Antioxid. Redox Signal. 14, 1093–1106 (2011).

    Article  CAS  PubMed  Google Scholar 

  41. Wang, Y., Yang, J. & Yi, J. Redox sensing by proteins: oxidative modifications on cysteines and the consequent events. Antioxid. Redox Signal. 16, 649–657 (2012).

    Article  PubMed  CAS  Google Scholar 

  42. Fu, X., Kassim, S. Y., Parks, W. C. & Heinecke, J. W. Hypochlorous acid oxygenates the cysteine switch domain of pro-matrilysin (MMP-7). A mechanism for matrix metalloproteinase activation and atherosclerotic plaque rupture by myeloperoxidase. J. Biol. Chem. 276, 41279–41287 (2001).

    Article  CAS  PubMed  Google Scholar 

  43. Taggart, C. et al. Oxidation of either methionine 351 or methionine 358 in α1-antitrypsin causes loss of anti-neutrophil elastase activity. J. Biol. Chem. 275, 27258–27265 (2000).

    CAS  PubMed  Google Scholar 

  44. Reddy, V. Y. et al. Oxidative dissociation of human α2-macroglobulin tetramers into dysfunctional dimers. J. Biol. Chem. 269, 4683–4691 (1994).

    CAS  PubMed  Google Scholar 

  45. Carp, H. & Janoff, A. Inactivation of bronchial mucous proteinase inhibitor by cigarette smoke and phagocyte-derived oxidants. Exp. Lung Res. 1, 225–237 (1980).

    Article  CAS  PubMed  Google Scholar 

  46. Lando, D., Peet, D. J., Whelan, D. A., Gorman, J. J. & Whitelaw, M. L. Asparagine hydroxylation of the HIF transactivation domain a hypoxic switch. Science 295, 858–861 (2002).

    Article  CAS  PubMed  Google Scholar 

  47. Doucette, C. D. Schwab, D.J., Wingreen, N. S. & Rabinowitz, J. D. α-Ketoglutarate coordinates carbon and nitrogen utilization via enzyme I inhibition. Nature Chem. Biol. 7, 894–901 (2011).

    Article  CAS  Google Scholar 

  48. Leichert, L. I. & Jakob, U. Protein thiol modifications visualized in vivo. PLoS Biol. 2, e333 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. White, A. A., Crawford, K. M., Patt, C. S. & Lad, P. J. Activation of soluble guanylate cyclase from rat lung by incubation or by hydrogen peroxide. J. Biol. Chem. 251, 7304–7312 (1976).

    CAS  PubMed  Google Scholar 

  50. Feng, W., Liu, G., Allen, P. D. & Pessah, I. N. Transmembrane redox sensor of ryanodine receptor complex. J. Biol. Chem. 275, 35902–35907 (2000).

    Article  CAS  PubMed  Google Scholar 

  51. Karisch, R. et al. Global proteomic assessment of the classical protein-tyrosine phosphatome and “Redoxome”. Cell 146, 826–840 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Finkel, T. Signal transduction by reactive oxygen species. J. Cell Biol. 194, 7–15 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Cosentino, C., Grieco, D. & Costanzo, V. ATM activates the pentose phosphate pathway promoting anti-oxidant defence and DNA repair. EMBO J. 30, 546–555 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Ito, K. et al. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nature Med. 12, 446–451 (2006).

    Article  CAS  PubMed  Google Scholar 

  55. Okuno, Y., Nakamura-Ishizu, A., Otsu, K., Suda, T. & Kubota, Y. Pathological neoangiogenesis depends on oxidative stress regulation by ATM. Nature Med. 18, 1208–1216 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Storz, G., Tartaglia, L. A. & Ames, B. N. Transcriptional regulator of oxidative stress-inducible genes: direct activation by oxidation. Science 248, 189–194 (1990).

    Article  CAS  PubMed  Google Scholar 

  57. Brunelle, J. K. et al. Oxygen sensing requires mitochondrial ROS but not oxidative phosphorylation. Cell. Metab. 1, 409–414 (2005).

    Article  CAS  PubMed  Google Scholar 

  58. Guzy, R. D. et al. Mitochondrial complex III is required for hypoxia-induced ROS production and cellular oxygen sensing. Cell. Metab. 1, 401–408 (2005).

    Article  CAS  PubMed  Google Scholar 

  59. Mansfield, K. D. et al. Mitochondrial dysfunction resulting from loss of cytochrome c impairs cellular oxygen sensing and hypoxic HIF-α activation. Cell. Metab. 1, 393–399 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ruchko, M. V. et al. Hypoxia-induced oxidative base modifications in the VEGF hypoxia-response element are associated with transcriptionally active nucleosomes. Free Radic. Biol. Med. 46, 352–359 (2009).

    Article  CAS  PubMed  Google Scholar 

  61. Al-Mehdi, A. B. et al. Perinuclear mitochondrial clustering creates an oxidant-rich nuclear domain required for hypoxia-induced transcription. Sci. Signal. 5, ra47 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Ju, B. G. et al. A topoisomerase IIβ-mediated dsDNA break required for regulated transcription. Science 312, 1798–1802 (2006).

    Article  CAS  PubMed  Google Scholar 

  63. Perillo, B. et al. DNA oxidation as triggered by H3K9me2 demethylation drives estrogen-induced gene expression. Science 319, 202–206 (2008).

    Article  CAS  PubMed  Google Scholar 

  64. Amente, S., Lania, L., Avvedimento, E. V. & Majello, B. DNA oxidation drives Myc mediated transcription. Cell Cycle 9, 3002–3004 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. Niethammer, P., Grabher, C., Look, A. T. & Mitchison, T. J. A tissue-scale gradient of hydrogen peroxide mediates rapid wound detection in zebrafish. Nature 459, 996–999 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Yoo, S. K., Starnes, T. W., Deng, Q. & Huttenlocher, A. Lyn is a redox sensor that mediates leukocyte wound attraction in vivo. Nature 480, 109–112 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Sakai, D. et al. Remodeling of actin cytoskeleton in mouse periosteal cells under mechanical loading induces periosteal cell proliferation during bone formation. PLoS ONE 6, e24847 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Hattori, H. et al. Small-molecule screen identifies reactive oxygen species as key regulators of neutrophil chemotaxis. Proc. Natl Acad. Sci. USA 107, 3546–3551 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Henderson, W. R. & Klebanoff, S. J. Leukotriene production and inactivation by normal, chronic granulomatous disease and myeloperoxidase-deficient neutrophils. J. Biol. Chem. 258, 13522–13527 (1983).

    CAS  PubMed  Google Scholar 

  70. Segal, B. H., Kuhns, D. B., Ding, L., Gallin, J. I. & Holland, S. M. Thioglycollate peritonitis in mice lacking C5, 5-lipoxygenase, or p47phox: complement, leukotrienes, and reactive oxidants in acute inflammation. J. Leukoc. Biol. 71, 410–416 (2002).

    CAS  PubMed  Google Scholar 

  71. Martinon, F., Chen, X., Lee, A. H. & Glimcher, L. H. TLR activation of the transcription factor XBP1 regulates innate immune responses in macrophages. Nature Immunol. 11, 411–418 (2010).

    Article  CAS  Google Scholar 

  72. West, A. P. et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature 472, 476–480 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Zhou, R., Yazdi, A. S., Menu, P. & Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 469, 221–225 (2011).

    Article  CAS  PubMed  Google Scholar 

  74. Shimada, K. et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 36, 401–414 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Dostert, C. et al. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 320, 674–677 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Hogquist, K. A., Nett, M. A., Unanue, E. R. & Chaplin, D. D. Interleukin 1 is processed and released during apoptosis. Proc. Natl Acad. Sci. USA 88, 8485–8489 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Shiloh, M. U. et al. Phenotype of mice and macrophages deficient in both phagocyte oxidase and inducible nitric oxide synthase. Immunity 10, 29–38 (1999).

    Article  CAS  PubMed  Google Scholar 

  78. Huang, J. et al. Activation of antibacterial autophagy by NADPH oxidases. Proc. Natl Acad. Sci. USA 106, 6226–6231 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Espey, M. G. Role of oxygen gradients in shaping redox relationships between the human intestine and its microbiota. Free Radic. Biol. Med. 55, 130–140 (2013).

    Article  CAS  PubMed  Google Scholar 

  80. Kumar, A. et al. Commensal bacteria modulate cullin-dependent signaling via generation of reactive oxygen species. EMBO J. 26, 4457–4466 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Winter, S. E. et al. Gut inflammation provides a respiratory electron acceptor for Salmonella. Nature 467, 426–429 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Maly, F. E. et al. The superoxide generating system of B cell lines. Structural homology with the phagocytic oxidase and triggering via surface Ig. J. Immunol. 140, 2334–2339 (1988).

    CAS  PubMed  Google Scholar 

  83. Maly, F. E. et al. Superoxide-dependent nitroblue tetrazolium reduction and expression of cytochrome b-245 components by human tonsillar B lymphocytes and B cell lines. J. Immunol. 142, 1260–1267 (1989).

    CAS  PubMed  Google Scholar 

  84. Singh, D. K. et al. The strength of receptor signaling is centrally controlled through a cooperative loop between Ca2+ and an oxidant signal. Cell 121, 281–293 (2005).

    Article  CAS  PubMed  Google Scholar 

  85. Devadas, S., Zaritskaya, L., Rhee, S. G., Oberley, L. & Williams, M. S. Discrete generation of superoxide and hydrogen peroxide by T cell receptor stimulation: selective regulation of mitogen-activated protein kinase activation and fas ligand expression. J. Exp. Med. 195, 59–70 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Hara-Chikuma, M. et al. Chemokine-dependent T cell migration requires aquaporin-3-mediated hydrogen peroxide uptake. J. Exp. Med. 209 1743–1752 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Los, M. et al. IL-2 gene expression and NF-kappa B activation through CD28 requires reactive oxygen production by 5-lipoxygenase. EMBO J. 14, 3731–3740 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Savina, A. et al. NOX2 controls phagosomal pH to regulate antigen processing during crosspresentation by dendritic cells. Cell 126, 205–218 (2006).

    Article  CAS  PubMed  Google Scholar 

  89. Mishell, R. I. & Dutton, R. W. Immunization of normal mouse spleen cell suspensions in vitro. Science 153, 1004–1006 (1966).

    Article  CAS  PubMed  Google Scholar 

  90. Nathan, C. F. & Terry, W. D. Differential stimulation of murine lymphoma growth in vitro by normal and BCG-activated macrophages. J. Exp. Med. 142, 887–902 (1975).

    Article  CAS  PubMed  Google Scholar 

  91. Angelini, G. et al. Antigen-presenting dendritic cells provide the reducing extracellular microenvironment required for T lymphocyte activation. Proc. Natl Acad. Sci. USA 99, 1491–1496 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Sido, B. et al. A prominent role for mucosal cystine/cysteine metabolism in intestinal immunoregulation. Gastroenterology 134, 179–191 (2008).

    Article  CAS  PubMed  Google Scholar 

  93. Yan, Z., Garg, S. K., Kipnis, J. & Banerjee, R. Extracellular redox modulation by regulatory T cells. Nature Chem. Biol. 5, 721–723 (2009).

    Article  CAS  Google Scholar 

  94. Fisher, R. I. & Bostick-Bruton, F. Depressed T cell proliferative responses in Hodgkin's disease: role of monocyte-mediated suppression via prostaglandins and hydrogen peroxide. J. Immunol. 129, 1770–1774 (1982).

    CAS  PubMed  Google Scholar 

  95. Efimova, O., Szankasi, P. & Kelley, T. W. Ncf1 (p47phox) is essential for direct regulatory T cell mediated suppression of CD4+ effector T cells. PLoS ONE 6, e16013 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Gelderman, K. A., Hultqvist, M., Holmberg, J., Olofsson, P. & Holmdahl, R. T cell surface redox levels determine T cell reactivity and arthritis susceptibility. Proc. Natl Acad. Sci. USA 103, 12831–12836 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Mougiakakos, D., Johansson, C. C., Jitschin, R., Bottcher, M. & Kiessling, R. Increased thioredoxin-1 production in human naturally occurring regulatory T cells confers enhanced tolerance to oxidative stress. Blood 117, 857–861 (2011).

    Article  CAS  PubMed  Google Scholar 

  98. Colombo, M. P. & Piconese, S. Regulatory-T-cell inhibition versus depletion: the right choice in cancer immunotherapy. Nature Rev. Cancer 7, 880–887 (2007).

    Article  CAS  Google Scholar 

  99. Gabrilovich, D. I. & Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nature Rev. Immunol. 9, 162–174 (2009).

    Article  CAS  Google Scholar 

  100. Muhlebach, T. J. et al. Treatment of patients with chronic granulomatous disease with recombinant human interferon-gamma does not improve neutrophil oxidative metabolism, cytochrome b558 content or levels of four anti-microbial proteins. Clin. Exp. Immunol. 88, 203–206 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Nagaraj, S. et al. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nature Med. 13, 828–835 (2007).

    CAS  PubMed  Google Scholar 

  102. Kusmartsev, S. & Gabrilovich, D. I. Inhibition of myeloid cell differentiation in cancer: the role of reactive oxygen species. J. Leuk. Biol. 74, 186–196 (2003).

    Article  CAS  Google Scholar 

  103. Molon, B. et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 208, 1949–1962 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Nathan, C. & Cohn, Z. Role of oxygen-dependent mechanisms in antibody-induced lysis of tumor cells by activated macrophages. J. Exp. Med. 152, 198–208 (1980).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Nathan, C. F. & Klebanoff, S. J. Augmentation of spontaneous macrophage-mediated cytolysis by eosinophil peroxidase. J. Exp. Med. 155, 1291–1308 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Nathan, C. F., Silverstein, S. C., Brukner, L. H. & Cohn, Z. A. Extracellular cytolysis by activated macrophages and granulocytes. II. Hydrogen peroxide as a mediator of cytotoxicity. J. Exp. Med. 149, 100–113 (1979).

    Article  CAS  PubMed  Google Scholar 

  107. Nathan, C. F., Arrick, B. A., Murray, H. W., DeSantis, N. M. & Cohn, Z. A. Tumor cell anti-oxidant defenses. Inhibition of the glutathione redox cycle enhances macrophage-mediated cytolysis. J. Exp. Med. 153, 766–782 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Nathan, C. F. & Cohn, Z. A. Antitumor effects of hydrogen peroxide in vivo. J. Exp. Med. 154, 1539–1553 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. O'Donnell-Tormey, J., DeBoer, C. J. & Nathan, C. F. Resistance of human tumor cells in vitro to oxidative cytolysis. J. Clin. Invest. 76, 80–86 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Szatrowski, T. P. & Nathan, C. F. Production of large amounts of hydrogen peroxide by human tumor cells. Cancer Res. 51, 794–798 (1991).

    CAS  PubMed  Google Scholar 

  111. Liou, G. Y. & Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 44, 479–496 (2010).

    Article  CAS  PubMed  Google Scholar 

  112. Ishikawa, K. et al. ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis. Science 320, 661–664 (2008).

    Article  CAS  PubMed  Google Scholar 

  113. Tonks, N. K. Redox redux: revisiting PTPs and the control of cell signaling. Cell 121, 667–670 (2005).

    Article  CAS  PubMed  Google Scholar 

  114. Ward, P. S. & Thompson, C. B. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell 21, 297–308 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Weitzman, S. A., Weitberg, A. B., Clark, E. P. & Stossel, T. P. Phagocytes as carcinogens: malignant transformation produced by human neutrophils. Science 227, 1231–1233 (1985).

    Article  CAS  PubMed  Google Scholar 

  116. Lonkar, P. & Dedon, P. C. Reactive species and DNA damage in chronic inflammation: reconciling chemical mechanisms and biological fates. Int. J. Cancer 128, 1999–2009 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Kohanski, M. A., DePristo, M. A. & Collins, J. J. Sublethal antibiotic treatment leads to multidrug resistance via radical-induced mutagenesis. Mol. Cell 37, 311–320 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Ishimoto, T. et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc and thereby promotes tumor growth. Cancer Cell 19, 387–400 (2011).

    Article  CAS  PubMed  Google Scholar 

  119. Gilbertson, R. J. & Rich, J. N. Making a tumour's bed: glioblastoma stem cells and the vascular niche. Nature Rev. Cancer 7, 733–736 (2007).

    Article  CAS  Google Scholar 

  120. Diehn, M. et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 458, 780–783 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Knoefler, D. et al. Quantitative in vivo redox sensors uncover oxidative stress as an early event in life. Mol. Cell 47, 767–776 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Gomes, A., Fernandes, E. & Lima, J. L. Fluorescence probes used for detection of reactive oxygen species. J. Biochem. Biophys. Methods 65, 45–80 (2005).

    Article  CAS  PubMed  Google Scholar 

  123. Kim, J. H. et al. Single-molecule detection of H2O2 mediating angiogenic redox signaling on fluorescent single-walled carbon nanotube array. ACS Nano 5, 7848–7857 (2011).

    Article  CAS  PubMed  Google Scholar 

  124. Lee, D. et al. Detection of hydrogen peroxide with chemiluminescent micelles. Int. J. Nanomed. 3, 471–476 (2008).

    Article  CAS  Google Scholar 

  125. Belousov, V. V. et al. Genetically encoded fluorescent indicator for intracellular hydrogen peroxide. Nature Methods 3, 281–286 (2006).

    Article  CAS  PubMed  Google Scholar 

  126. Gutscher, M. et al. Real-time imaging of the intracellular glutathione redox potential. Nature Methods 5, 553–559 (2008).

    Article  CAS  PubMed  Google Scholar 

  127. Raj, L. et al. Selective killing of cancer cells by a small molecule targeting the stress response to ROS. Nature 475, 231–234 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Trachootham, D., Alexandre, J. & Huang, P. Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nature Rev. Drug Discov. 8, 579–591 (2009).

    Article  CAS  Google Scholar 

  129. Kohanski, M. A., Dwyer, D. J., Hayete, B., Lawrence, C. A. & Collins, J. J. A common mechanism of cellular death induced by bactericidal antibiotics. Cell 130, 797–810 (2007).

    Article  CAS  PubMed  Google Scholar 

  130. Foti, J. J., Devadoss, B., Winkler, J. A., Collins, J. J. & Walker, G. C. Oxidation of the guanine nucleotide pool underlies cell death by bactericidal antibiotics. Science 336, 315–319 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Nathan, C. Fresh approaches to anti-infective therapies. Sci. Transl. Med. 4, 140sr2 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Liby, K. T. & Sporn, M. B. Synthetic oleanane triterpenoids: multifunctional drugs with a broad range of applications for prevention and treatment of chronic disease. Pharmacol. Rev. 64, 972–1003 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Pineda-Molina, E. et al. Glutathionylation of the p50 subunit of NF-κB: a mechanism for redox-induced inhibition of DNA binding. Biochemistry 40, 14134–14142 (2001).

    Article  CAS  PubMed  Google Scholar 

  134. Warburg, O. Beobachtungen über die Oxydationsprozesse im Seeigelei. Z. Physiol. Chem. 57, 1–16 (1908).

    Article  Google Scholar 

  135. Bentley, R. in The Enzymes 2nd edn Vol. 27 Ch. 24 (eds Boyer, P. D., Lardy, H. & Myrbäck, K.) 567–586 (Academic, 1963).

    Google Scholar 

  136. Wilson, R. & Turner, A. P. F. Glucose oxidase: an ideal enzyme. Biosensors & Bioelectronics 7, 165–185 (1992).

    Article  CAS  Google Scholar 

  137. Sbarra, A. J. & Karnovsky, M. L. The biochemical basis of phagocytosis. I. Metabolic changes during the ingestion of particles by polymorphonuclear leukocytes. J. Biol. Chem. 234, 1355–1362 (1959).

    CAS  PubMed  Google Scholar 

  138. Iyer, G. Y. N., Islam, M. F. & Quastel, J. H. Biochemical aspects of phagocytosis. Nature 192, 535–541 (1961).

    Article  CAS  Google Scholar 

  139. McCord, J. M. & Fridovich, I. Superoxide dismutase. An enzymic function for erythrocuprein (hemocuprein). J. Biol. Chem. 244, 6049–6055 (1969).

    CAS  PubMed  Google Scholar 

  140. Babior, B. M., Kipnes, R. S. & Curnutte, J. T. Biological defense mechanisms. The production by leukocytes of superoxide, a potential bactericidal agent. J. Clin. Invest. 52, 741–744 (1973).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Curnutte, J. T., Whitten, D. M. & Babior, B. M. Defective superoxide production by granulocytes from patients with chronic granulomatous disease. N. Engl. J. Med. 290, 593–597 (1974).

    Article  CAS  PubMed  Google Scholar 

  142. Nathan, C. F. & Root, R. K. Hydrogen peroxide release from mouse peritoneal macrophages: dependence on sequential activation and triggering. J. Exp. Med. 146, 1648–1662 (1977).

    Article  CAS  PubMed  Google Scholar 

  143. Foerder, C. A., Klebanoff, S. J. & Shapiro, B. M. Hydrogen peroxide production, chemiluminescence, and the respiratory burst of fertilization: interrelated events in early sea urchin development. Proc. Natl Acad. Sci. USA 75, 3183–3187 (1978).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Segal, A. W. & Jones, O. T. Novel cytochrome b system in phagocytic vacuoles of human granulocytes. Nature 276, 515–517 (1978).

    Article  CAS  PubMed  Google Scholar 

  145. Klebanoff, S. J. Oxygen metabolism and the toxic properties of phagocytes. Ann. Intern. Med. 93, 480–489 (1980).

    Article  CAS  PubMed  Google Scholar 

  146. Nathan, C. F., Murray, H. W., Wiebe, M. E. & Rubin, B. Y. Identification of interferon-gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J. Exp. Med. 158, 670–689 (1983).

    Article  CAS  PubMed  Google Scholar 

  147. Nathan, C. F. et al. Local and systemic effects of intradermal recombinant interferon-γ in patients with lepromatous leprosy. N. Engl. J. Med. 315, 6–15 (1986).

    Article  CAS  PubMed  Google Scholar 

  148. Royer-Pokora, B. et al. Cloning the gene for an inherited human disorder — chronic granulomatous disease — on the basis of its chromosomal location. Nature 322, 32–38 (1986).

    Article  CAS  PubMed  Google Scholar 

  149. Ezekowitz, R. A., Dinauer, M. C., Jaffe, H. S., Orkin, S. H. & Newburger, P. E. Partial correction of the phagocyte defect in patients with X-linked chronic granulomatous disease by subcutaneous interferon gamma. N. Engl. J. Med. 319, 146–151 (1988).

    Article  CAS  PubMed  Google Scholar 

  150. Suh, Y. A. et al. Cell transformation by the superoxide-generating oxidase Mox1. Nature 401, 79–82 (1999).

    Article  CAS  PubMed  Google Scholar 

  151. Grant, S. S., Kaufmann, B. B., Chand, N. S., Haseley, N. & Hung, D. T. Eradication of bacterial persisters with antibiotic-generated hydroxyl radicals. Proc. Natl Acad. Sci. USA 109, 12147–12152 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Doroshow, J. H. & Davies, K. J. Comparative cardiac oxygen radical metabolism by anthracycline antibiotics, mitoxantrone, bisantrene, 4′-(9-acridinylamino)-methanesulfon-m-anisidide, and neocarzinostatin. Biochem. Pharmacol. 32, 2935–2939 (1983).

    Article  CAS  PubMed  Google Scholar 

  153. Dorr, R.T. Bleomycin pharmacology: mechanism of action and resistance, and clinical pharmacokinetics. Semin. Oncol. 19, 3–8 (1992).

    CAS  PubMed  Google Scholar 

  154. Liu, Y. & Imlay, J. A. Cell death from antibiotics without the involvement of reactive oxygen species. Science 339, 1210 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Keren, I., Wu, Y., Inocencio, U., Mulcahy, L. R. & Lewis, K. Killing by bactericidal antibiotics does not depend on reactive oxygen species. Science 339, 1213 (2013).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

A.C.-B. is a member of the Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Programme, which is supported by the Medical Scientist Training Program grant (GM07739) from the National Instiute of General Medical Sciences, USA. The Department of Microbiology and Immunology is supported by the William Randolph Hearst Trust.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carl Nathan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Carl Nathan's homepage

Glossary

Iron–sulphur clusters

Prosthetic groups that are required for the function of some enzymes. In iron–sulphur clusters two, three or four atoms of iron are attached to the protein through two or four sulphydryl groups.

Uncoupling proteins

Proteins in the mitochondrial inner membrane that can divert the proton gradient away from the formation of ATP, resulting in the generation of heat instead.

Xenobiotics

Small chemical compounds that enter an organism unnaturally, such as drugs or pollutants.

Acidic dissociation constant

(pKa). The equilibrium constant for the dissociation of an acid into its conjugate base and hydrogen ion, expressed as the negative logarithm. The lower the pKa of a sulphydryl group, the greater the likelihood that the sulphur will be anionic at ambient pH.

Chronic granulomatous disease

(CGD). An immunodeficiency state manifested by recurrent, often life-threatening, infections and the excessive formation of granulomas, caused by mutations in any one of four subunits of NADP oxidase 2.

Granulomas

Histological collections of macrophages, usually surrounded by lymphocytes and sometimes fibrocytes. Some of the macrophages might seem to be 'epithelioid' or fuse to become multinucleated giant cells. Granuloma formation is a chronic inflammatory response to various infectious and non-infectious agents.

Neddylation

A process that is analogous to ubiquitylation, in which ubiquitin-like protein NEDD8 is conjugated to a protein substrate.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nathan, C., Cunningham-Bussel, A. Beyond oxidative stress: an immunologist's guide to reactive oxygen species. Nat Rev Immunol 13, 349–361 (2013). https://doi.org/10.1038/nri3423

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri3423

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer