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:

Fundamentals of redox regulation in biology

Abstract

Oxidation–reduction (redox) reactions are central to the existence of life. Reactive species of oxygen, nitrogen and sulfur mediate redox control of a wide range of essential cellular processes. Yet, excessive levels of oxidants are associated with ageing and many diseases, including cardiological and neurodegenerative diseases, and cancer. Hence, maintaining the fine-tuned steady-state balance of reactive species production and removal is essential. Here, we discuss new insights into the dynamic maintenance of redox homeostasis (that is, redox homeodynamics) and the principles underlying biological redox organization, termed the ‘redox code’. We survey how redox changes result in stress responses by hormesis mechanisms, and how the lifelong cumulative exposure to environmental agents, termed the ‘exposome’, is communicated to cells through redox signals. Better understanding of the molecular and cellular basis of redox biology will guide novel redox medicine approaches aimed at preventing and treating diseases associated with disturbed redox regulation.

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

Fig. 1: Principles of redox regulation.
Fig. 2: Redox eustress and transition to oxidative and reductive distress.
Fig. 3: Schematic overview of redox communication in and between cells.
Fig. 4: Major cellular processes under redox regulation.

Similar content being viewed by others

References

  1. Walsh, C. T., Tu, B. P. & Tang, Y. Eight kinetically stable but thermodynamically activated molecules that power cell metabolism. Chem. Rev. 118, 1460–1494 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Jacob, C., Giles, G. I., Giles, N. M. & Sies, H. Sulfur and selenium: the role of oxidation state in protein structure and function. Angew. Chem. Int. Ed. Engl. 42, 4742–4758 (2003).

    Article  CAS  PubMed  Google Scholar 

  3. Lennicke, C. & Cochemé, H. M. Redox metabolism: ROS as specific molecular regulators of cell signaling and function. Mol. Cell 81, 3691–3707 (2021).

    Article  CAS  PubMed  Google Scholar 

  4. Sies, H. et al. Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat. Rev. Mol. Cell Biol. 23, 499–515 (2022). This ‘Expert Recommendation’ addresses key questions regarding the impact of oxidants on physiology and their contribution to disease.

    Article  CAS  PubMed  Google Scholar 

  5. Thannickal, V. J. & Fanburg, B. L. Reactive oxygen species in cell signaling. Am. J. Physiol. Lung Cell Mol. Physiol. 279, L1005–L1028 (2000).

    Article  CAS  PubMed  Google Scholar 

  6. D’Autreaux, B. & Toledano, M. B. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 8, 813–824 (2007). This article describes fundamental perspectives on reactive oxygen species signalling.

    Article  PubMed  Google Scholar 

  7. Butterfield, D. A. & Perluigi, M. Redox proteomics: a key tool for new insights into protein modification with relevance to disease. Antioxid. Redox Signal. 26, 277–279 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Murphy, M. P. et al. Guidelines for measuring reactive oxygen species and oxidative damage in cells and in vivo. Nat. Metab. 4, 651–662 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Sies, H. & Jones, D. P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 21, 363–383 (2020).

    Article  CAS  PubMed  Google Scholar 

  10. Radi, R. Oxygen radicals, nitric oxide, and peroxynitrite: redox pathways in molecular medicine. Proc. Natl Acad. Sci. USA 115, 5839–5848 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lundberg, J. O. & Weitzberg, E. Nitric oxide signaling in health and disease. Cell 185, 2853–2878 (2022).

    Article  CAS  PubMed  Google Scholar 

  12. Cirino, G., Szabo, C. & Papapetropoulos, A. Physiological roles of hydrogen sulfide in mammalian cells, tissues, and organs. Physiol. Rev. 103, 31–276 (2023).

    Article  CAS  PubMed  Google Scholar 

  13. Parvez, S., Long, M. J. C., Poganik, J. R. & Aye, Y. Redox signaling by reactive electrophiles and oxidants. Chem. Rev. 118, 8798–8888 (2018). This is a comprehensive review of the signalling role of electrophiles and oxidants in biology.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Rabbani, N. & Thornalley, P. J. Protein glycation — biomarkers of metabolic dysfunction and early-stage decline in health in the era of precision medicine. Redox Biol. 42, 101920 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Noctor, G. & Foyer, C. H. Intracellular redox compartmentation and ROS-related communication in regulation and signaling. Plant Physiol. 171, 1581–1592 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mittler, R., Zandalinas, S. I., Fichman, Y. & Van Breusegem, F. Reactive oxygen species signalling in plant stress responses. Nat. Rev. Mol. Cell Biol. 23, 663–679 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Dietz, K. J. & Vogelsang, L. A general concept of quantitative abiotic stress sensing. Trends Plant Sci. 23, 237–246 (2023).

    Google Scholar 

  18. Sies, H. (ed.) Oxidative Stress: Eustress and Distress 1–844 (Academic Press, 2020).

  19. Wild, C. P. The exposome: from concept to utility. Int. J. Epidemiol. 41, 24–32 (2012).

    Article  PubMed  Google Scholar 

  20. Xiao, W. & Loscalzo, J. Metabolic responses to reductive stress. Antioxid. Redox Signal. 32, 1330–1347 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hanschmann, E. M., Godoy, J. R., Berndt, C., Hudemann, C. & Lillig, C. H. Thioredoxins, glutaredoxins, and peroxiredoxins — molecular mechanisms and health significance: from cofactors to antioxidants to redox signaling. Antioxid. Redox Signal. 19, 1539–1605 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Jones, D. P. & Sies, H. The redox code. Antioxid. Redox Signal. 23, 734–746 (2015). This article introduces the ‘redox code’ as a set of fundamental principles of organization of biological redox reactions.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Prigogine, I. Time, structure, and fluctuations. Science 201, 777–785 (1978).

    Article  CAS  PubMed  Google Scholar 

  24. Sies, H. Oxidative eustress: on constant alert for redox homeostasis. Redox Biol. 41, 101867 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kondadi, A. K. et al. Cristae undergo continuous cycles of membrane remodelling in a MICOS-dependent manner. EMBO Rep. 21, e49776 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Billman, G. E. Homeostasis: the underappreciated and far too often ignored central organizing principle of physiology. Front. Physiol. 11, 200 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Lloyd, D., Aon, M. A. & Cortassa, S. Why homeodynamics, not homeostasis? Scientific World J. 1, 133–145 (2001).

    Article  CAS  Google Scholar 

  28. Xiong, L. I. & Garfinkel, A. Are physiological oscillations physiological? J. Physiol. https://doi.org/10.1113/JP285015 (2023).

    Article  PubMed  Google Scholar 

  29. Brash, D. E. Rethinking causation for data-intensive biology: constraints, cancellations, and quantized organisms: causality in complex organisms is sculpted by constraints rather than instigators, with outcomes perhaps better described by quantized patterns than rectilinear pathways. Bioessays 42, e1900135 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Sies, H., Berndt, C. & Jones, D. P. Oxidative stress. Annu. Rev. Biochem. 86, 715–748 (2017). This article discusses the concept of oxidative stress, physiological (eustress) and supraphysiological (distress).

    Article  CAS  PubMed  Google Scholar 

  31. Lushchak, V. I. & Storey, K. B. Oxidative stress concept updated: definitions, classifications and regulatory pathways implicated. EXCLI J. 20, 956–967 (2021).

    PubMed  PubMed Central  Google Scholar 

  32. Rattan, S. I. Molecular gerontology: from homeodynamics to hormesis. Curr. Pharm. Des. 20, 3036–3039 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Alleman, R. J., Katunga, L. A., Nelson, M. A., Brown, D. A. & Anderson, E. J. The ‘Goldilocks Zone’ from a redox perspective — adaptive vs. deleterious responses to oxidative stress in striated muscle. Front. Physiol. 5, 358 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Ursini, F., Maiorino, M. & Forman, H. J. Redox homeostasis: the golden mean of healthy living. Redox Biol. 8, 205–215 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sies, H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: oxidative eustress. Redox Biol. 11, 613–619 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Winterbourn, C. C. Are free radicals involved in thiol-based redox signaling? Free Radic. Biol. Med. 80, 164–170 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Holmström, K. M. & Finkel, T. Cellular mechanisms and physiological consequences of redox-dependent signalling. Nat. Rev. Mol. Cell Biol. 15, 411–421 (2014).

    Article  PubMed  Google Scholar 

  38. Brigelius-Flohé, R. & Flohé, L. Basic principles and emerging concepts in the redox control of transcription factors. Antioxid. Redox Signal. 15, 2335–2381 (2011). Together with Marinho et al. (2014), this comprehensive review discusses redox control of transcription factors.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Marinho, H. S., Real, C., Cyrne, L., Soares, H. & Antunes, F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2, 535–562 (2014). Together with Brigelius-Flohé and Flohé (2011), this comprehensive review discusses redox control of transcription factors.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Jose, E. et al. Temporal coordination of the transcription factor response to H2O2 stress. Nat. Commun. 15, 3440 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Müller, N. et al. Reactive oxygen species differentially modulate the metabolic and transcriptomic response of endothelial cells. Antioxidants 11, 434 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Suzuki, T. & Yamamoto, M. Stress-sensing mechanisms and the physiological roles of the Keap1-Nrf2 system during cellular stress. J. Biol. Chem. 292, 16817–16824 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Yamamoto, M., Kensler, T. W. & Motohashi, H. The KEAP1-nrf2 system: a thiol-based sensor-effector apparatus for maintaining redox homeostasis. Physiol. Rev. 98, 1169–1203 (2018). A comprehensive review of the mechanism, biology and medical aspects of the KEAP1–NRF2 system that regulates responses to oxidative and electrophilic challenges.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Cuadrado, A. et al. Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nat. Rev. Drug Discov. 18, 295–317 (2019).

    Article  CAS  PubMed  Google Scholar 

  45. Calabrese, E. J. & Kozumbo, W. J. The hormetic dose–response mechanism: Nrf2 activation. Pharmacol. Res. 167, 105526 (2021). This article describes the pivotal role of the NRF2 system in hormetic responses.

    Article  CAS  PubMed  Google Scholar 

  46. Agathokleous, E. & Calabrese, E. J. Hormesis: a general biological principle. Chem. Res. Toxicol. 35, 547–549 (2022).

    Article  CAS  PubMed  Google Scholar 

  47. Mattson, M. P. & Leak, R. K. The hormesis principle of neuroplasticity and neuroprotection. Cell Metab. 36, 315–337 (2024).

    Article  CAS  PubMed  Google Scholar 

  48. Baird, L. et al. A NRF2-induced secretory phenotype activates immune surveillance to remove irreparably damaged cells. Redox Biol. 66, 102845 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Agarwal, S. & Ganesh, S. Perinuclear mitochondrial clustering, increased ROS levels, and HIF1 are required for the activation of HSF1 by heat stress. J. Cell Sci. 133, jcs245589 (2020).

    Article  CAS  PubMed  Google Scholar 

  50. 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  Google Scholar 

  51. Cyran, A. M. & Zhitkovich, A. HIF1, HSF1, and NRF2: oxidant-responsive trio raising cellular defenses and engaging immune system. Chem. Res. Toxicol. 35, 1690–1700 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Liu, S. H., Qiu, Y., Xiang, R. & Huang, P. Characterization of H2O2-induced alterations in global transcription of mRNA and lncRNA. Antioxidants (Basel) 11, 495 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. D’Souza, L. C. et al. Oxidative stress and cancer development: are noncoding RNAs the missing links? Antioxid. Redox Signal. 33, 1209–1229 (2020).

    Article  PubMed  Google Scholar 

  54. Zheng, F. et al. The HIF-1alpha antisense long non-coding RNA drives a positive feedback loop of HIF-1alpha mediated transactivation and glycolysis. Nat. Commun. 12, 1341 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ji, Q., Zong, X., Mao, Y. & Qian, S. B. A heat shock-responsive lncRNA heat acts as a HSF1-directed transcriptional brake via m(6)A modification. Proc. Natl Acad. Sci. USA 118, e2102175118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Weiss-Sadan, T. et al. NRF2 activation induces NADH-reductive stress, providing a metabolic vulnerability in lung cancer. Cell Metab. 35, 722 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Manford, A. G. et al. A cellular mechanism to detect and alleviate reductive stress. Cell 183, 46–61.e21 (2020).

    Article  CAS  PubMed  Google Scholar 

  58. Galluzzi, L., Yamazaki, T. & Kroemer, G. Linking cellular stress responses to systemic homeostasis. Nat. Rev. Mol. Cell Biol. 19, 731–745 (2018).

    Article  CAS  PubMed  Google Scholar 

  59. Costa-Mattioli, M. & Walter, P. The integrated stress response: from mechanism to disease. Science 368, eaat5314 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hetz, C., Zhang, K. & Kaufman, R. J. Mechanisms, regulation and functions of the unfolded protein response. Nat. Rev. Mol. Cell Biol. 21, 421–438 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Lu, H., Yang, M. & Zhou, Q. Reprogramming transcription after DNA damage: recognition, response, repair, and restart. Trends Cell Biol. 33, 682–694 (2023).

    Article  CAS  PubMed  Google Scholar 

  62. Gonzalez-Quiroz, M. et al. When endoplasmic reticulum proteostasis meets the DNA damage response. Trends Cell Biol. 30, 881–891 (2020).

    Article  CAS  PubMed  Google Scholar 

  63. He, F., Ru, X. & Wen, T. NRF2, a transcription factor for stress response and beyond. Int. J. Mol. Sci. 21, 4777 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Johansson, K. et al. Cross talk in HEK293 cells between Nrf2, HIF, and NF-kappaB activities upon challenges with redox therapeutics characterized with single-cell resolution. Antioxid. Redox Signal. 26, 229–246 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Meng, J., Lv, Z., Wang, Y. & Chen, C. Identification of the redox-stress signaling threshold (RST): increased RST helps to delay aging in C. elegans. Free Radic. Biol. Med. 178, 54–58 (2022).

    Article  CAS  PubMed  Google Scholar 

  66. Pallepati, P. & Averill-Bates, D. A. Activation of ER stress and apoptosis by hydrogen peroxide in HeLa cells: protective role of mild heat preconditioning at 40 °C. Biochim. Biophys. Acta 1813, 1987–1999 (2011).

    Article  CAS  PubMed  Google Scholar 

  67. Pospelova, T. V. et al. Pseudo-DNA damage response in senescent cells. Cell Cycle 8, 4112–4118 (2009).

    Article  CAS  PubMed  Google Scholar 

  68. Halvey, P. J. et al. Compartmental oxidation of thiol-disulphide redox couples during epidermal growth factor signalling. Biochem. J. 386, 215–219 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Bergerhausen, L. et al. Extracellular redox regulation of α7β integrin-mediated cell migration is signaled via a dominant thiol-switch. Antioxidants 9, 227 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Nordzieke, D. E. & Medraño-Fernandez, I. The plasma membrane: a platform for intra- and intercellular redox signaling. Antioxidants 7, 168 (2018). This article provides a comprehensive overview of the plasma membrane as redox signalling platform.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Petersen, S. V., Poulsen, N. B., Linneberg Matthiesen, C. & Vilhardt, F. Novel and converging ways of NOX2 and SOD3 in trafficking and redox signaling in macrophages. Antioxidants 10, 172 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Bienert, G. P. & Chaumont, F. Aquaporin-facilitated transmembrane diffusion of hydrogen peroxide. Biochim. Biophys. Acta 1840, 1596–1604 (2014). This paper is a seminal work on the role of some aquaporins as peroxiporins.

    Article  CAS  PubMed  Google Scholar 

  73. Mishina, N. M. et al. Does cellular hydrogen peroxide diffuse or act locally? Antioxid. Redox Signal. 14, 1–7 (2011).

    Article  CAS  PubMed  Google Scholar 

  74. Oakley, F. D., Abbott, D., Li, Q. & Engelhardt, J. F. Signaling components of redox active endosomes: the redoxosomes. Antioxid. Redox Signal. 11, 1313–1333 (2009). These findings shaped the concept of redox-active endosomes as redoxosomes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Spencer, N. Y. & Engelhardt, J. F. The basic biology of redoxosomes in cytokine-mediated signal transduction and implications for disease-specific therapies. Biochemistry 53, 1551–1564 (2014).

    Article  CAS  PubMed  Google Scholar 

  76. Balta, E., Kramer, J. & Samstag, Y. Redox regulation of the actin cytoskeleton in cell migration and adhesion: on the way to a spatiotemporal view. Front. Cell Dev. Biol. 8, 618261 (2020).

    Article  PubMed  Google Scholar 

  77. Rouyère, C., Serrano, T., Fremont, S. & Echard, A. Oxidation and reduction of actin: origin, impact in vitro and functional consequences in vivo. Eur. J. Cell Biol. 101, 151249 (2022).

    Article  PubMed  Google Scholar 

  78. Kang, M. I., Kobayashi, A., Wakabayashi, N., Kim, S. G. & Yamamoto, M. Scaffolding of Keap1 to the actin cytoskeleton controls the function of Nrf2 as key regulator of cytoprotective phase 2 genes. Proc. Natl Acad. Sci. USA 101, 2046–2051 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Valdivia, A., Duran, C. & San Martin, A. The role of nox-mediated oxidation in the regulation of cytoskeletal dynamics. Curr. Pharm. Des. 21, 6009–6022 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Perez-Sala, D. & Quinlan, R. A. The redox-responsive roles of intermediate filaments in cellular stress detection, integration and mitigation. Curr. Opin. Cell Biol. 86, 102283 (2023).

    Article  PubMed  Google Scholar 

  81. Walker, C. L., Pomatto, L. C. D., Tripathi, D. N. & Davies, K. J. A. Redox regulation of homeostasis and proteostasis in peroxisomes. Physiol. Rev. 98, 89–115 (2018).

    Article  CAS  PubMed  Google Scholar 

  82. Fujiki, Y., Okumoto, K., Honsho, M. & Abe, Y. Molecular insights into peroxisome homeostasis and peroxisome biogenesis disorders. Biochim. Biophys. Acta Mol. Cell Res. 1869, 119330 (2022).

    Article  CAS  PubMed  Google Scholar 

  83. Sandalio, L. M., Collado-Arenal, A. M. & Romero-Puertas, M. C. Deciphering peroxisomal reactive species interactome and redox signalling networks. Free Radic. Biol. Med. 197, 58–70 (2023).

    Article  CAS  PubMed  Google Scholar 

  84. Lismont, C. et al. Peroxisome-derived hydrogen peroxide modulates the sulfenylation profiles of key redox signaling proteins in Flp-In T-REx 293 cells. Front. Cell Dev. Biol. 10, 888873 (2022). This work demonstrates the role of peroxisomal hydrogen peroxide in intracellular redox communication.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Okumoto, K. et al. The peroxisome counteracts oxidative stresses by suppressing catalase import via Pex14 phosphorylation. eLife 9, e55896 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Dubreuil, M. M. et al. Systematic identification of regulators of oxidative stress reveals non-canonical roles for peroxisomal import and the pentose phosphate pathway. Cell Rep. 30, 1417–1433 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Di Cara, F., Savary, S., Kovacs, W. J., Kim, P. & Rachubinski, R. A. The peroxisome: an up-and-coming organelle in immunometabolism. Trends Cell Biol. 33, 70–86 (2023).

    Article  PubMed  Google Scholar 

  88. Wenzel, E. M., Elfmark, L. A., Stenmark, H. & Raiborg, C. ER as master regulator of membrane trafficking and organelle function. J. Cell Biol. 221, e202205135 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Rashdan, N. A. & Pattillo, C. B. Hydrogen peroxide in the ER: a tale of triage. Redox Biol. 28, 101358 (2020).

    Article  CAS  PubMed  Google Scholar 

  90. Kirstein, J. et al. Proteotoxic stress and ageing triggers the loss of redox homeostasis across cellular compartments. EMBO J. 34, 2334–2349 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Jacobs, L. J. & Riemer, J. Maintenance of small molecule redox homeostasis in mitochondria. FEBS Lett. 597, 205–223 (2022).

    Article  PubMed  Google Scholar 

  92. Shadel, G. S. & Horvath, T. L. Mitochondrial ROS signaling in organismal homeostasis. Cell 163, 560–569 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Kondadi, A. K. & Reichert, A. S. Mitochondrial dynamics at different levels: from cristae dynamics to interorganellar cross talk. Annu. Rev. Biophys. https://doi.org/10.1146/annurev-biophys-030822-020736 (2024).

  94. Chakrabarty, R. P. & Chandel, N. S. Beyond ATP, new roles of mitochondria. Biochem 44, 2–8 (2022).

    Article  CAS  Google Scholar 

  95. Monzel, A. S., Enriquez, J. A. & Picard, M. Multifaceted mitochondria: moving mitochondrial science beyond function and dysfunction. Nat. Metab. 5, 546–562 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  96. Popov, L. D. Mitochondria as intracellular signalling organelles. An update. Cell Signal. 109, 110794 (2023).

    Article  CAS  PubMed  Google Scholar 

  97. Murphy, M. P. How mitochondria produce reactive oxygen species. Biochem. J. 417, 1–13 (2009).

    Article  CAS  PubMed  Google Scholar 

  98. Hoehne, M. N. et al. Spatial and temporal control of mitochondrial H2O2 release in intact human cells. EMBO J. 41, e109169 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Koren, S. A. et al. All-optical spatiotemporal mapping of ROS dynamics across mitochondrial microdomains in situ. Nat. Commun. 14, 6036 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Grayson, C. & Mailloux, R. J. Coenzyme Q(10) and nicotinamide nucleotide transhydrogenase: sentinels for mitochondrial hydrogen peroxide signaling. Free Radic. Biol. Med. 208, 260–271 (2023).

    Article  CAS  PubMed  Google Scholar 

  101. Mills, E. L. et al. Cysteine 253 of UCP1 regulates energy expenditure and sex-dependent adipose tissue inflammation. Cell Metab. 34, 140–157 (2022).

    Article  CAS  PubMed  Google Scholar 

  102. Pei, J. F. et al. Diurnal oscillations of endogenous H(2)O(2) sustained by p66(Shc) regulate circadian clocks. Nat. Cell Biol. 21, 1553–1564 (2019). This work demonstrates the circadian rhythm of endogenously produced hydrogen peroxide.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Funato, Y. et al. Nucleoredoxin, a novel thioredoxin family member involved in cell growth and differentiation. Antioxid. Redox Signal. 9, 1035–1057 (2007).

    Article  CAS  PubMed  Google Scholar 

  104. Idelfonso-Garcia, O. G. et al. Is nucleoredoxin a master regulator of cellular redox homeostasis? Its implication in different pathologies. Antioxidants 11, 670 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Seifermann, M. & Epe, B. Oxidatively generated base modifications in DNA: not only carcinogenic risk factor but also regulatory mark? Free Radic. Biol. Med. 107, 258–265 (2017).

    Article  CAS  PubMed  Google Scholar 

  106. Fleming, A. M. & Burrows, C. J. Chemistry of ROS-mediated oxidation to the guanine base in DNA and its biological consequences. Int. J. Radiat. Biol. 98, 452–460 (2022).

    Article  CAS  PubMed  Google Scholar 

  107. Hahm, J. Y., Park, J., Jang, E. S. & Chi, S. W. 8-Oxoguanine: from oxidative damage to epigenetic and epitranscriptional modification. Exp. Mol. Med. 54, 1626–1642 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Perillo, B., Tramontano, A., Pezone, A. & Migliaccio, A. LSD1: more than demethylation of histone lysine residues. Exp. Mol. Med. 52, 1936–1947 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Leisegang, M. S., Schröder, K. & Brandes, R. P. Redox regulation and noncoding RNAs. Antioxid. Redox Signal. 29, 793–812 (2018). This paper shows a comprehensive overview of the role of non-coding RNAs in redox regulation.

    Article  CAS  PubMed  Google Scholar 

  110. Lettieri-Barbato, D., Aquilano, K., Punziano, C., Minopoli, G. & Faraonio, R. MicroRNAs, long non-coding RNAs, and circular RNAs in the redox control of cell senescence. Antioxidants (Basel) 11, 480 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Ciesielska, S., Slezak-Prochazka, I., Bil, P. & Rzeszowska-Wolny, J. Micro RNAs in regulation of cellular redox homeostasis. Int. J. Mol. Sci. 22, 6022 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Zhang, Y. et al. Circular RNAs in the regulation of oxidative stress. Front. Pharmacol. 12, 697903 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Mercatelli, N. et al. MiR-23-TrxR1 as a novel molecular axis in skeletal muscle differentiation. Sci. Rep. 7, 7219 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Fuschi, P. et al. Central role of the p53 pathway in the noncoding-RNA response to oxidative stress. Aging 9, 2559–2586 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Prinz, W. A., Toulmay, A. & Balla, T. The functional universe of membrane contact sites. Nat. Rev. Mol. Cell Biol. 21, 7–24 (2020).

    Article  CAS  PubMed  Google Scholar 

  116. Nieto-Garai, J. A. et al. Super-resolution microscopy to study interorganelle contact sites. Int. J. Mol. Sci. 23, 15354 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Fuentes-Lemus, E. & Davies, M. J. Effect of crowding, compartmentalization and nanodomains on protein modification and redox signaling — current state and future challenges. Free Radic. Biol. Med. 196, 81–92 (2023).

    Article  CAS  PubMed  Google Scholar 

  118. Yoboue, E. D., Sitia, R. & Simmen, T. Redox crosstalk at endoplasmic reticulum (ER) membrane contact sites (MCS) uses toxic waste to deliver messages. Cell Death Dis. 9, 331–0033 (2018). This paper describes the concept of endoplasmic reticulum–mitochondria–peroxisomes crosstalk: ‘redox triangle’.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Gordaliza-Alaguero, I., Canto, C. & Zorzano, A. Metabolic implications of organelle–mitochondria communication. EMBO Rep. 20, e47928 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Resende, R., Fernandes, T., Pereira, A. C., Marques, A. P. & Pereira, C. F. Endoplasmic reticulum–mitochondria contacts modulate reactive oxygen species-mediated signaling and oxidative stress in brain disorders: the key role of sigma-1 receptor. Antioxid. Redox Signal. 37, 758–780 (2022).

    Article  CAS  PubMed  Google Scholar 

  121. Bestetti, S. et al. Human aquaporin-11 guarantees efficient transport of H(2)O(2) across the endoplasmic reticulum membrane. Redox Biol. 28, 101326 (2020).

    Article  CAS  PubMed  Google Scholar 

  122. Sorrentino, I., Galli, M., Medrano-Fernandez, I. & Sitia, R. Transfer of H2O2 from mitochondria to the endoplasmic reticulum via aquaporin-11. Redox Biol. 55, 102410 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Fox, A. R. et al. Plasma membrane aquaporins interact with the endoplasmic reticulum resident VAP27 proteins at ER-PM contact sites and endocytic structures. N. Phytol. 228, 973–988 (2020).

    Article  CAS  Google Scholar 

  124. Azad, A. K. et al. Human aquaporins: functional diversity and potential roles in infectious and non-infectious diseases. Front. Genet. 12, 654865 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Balderas, P. M. D. Mitochondria–plasma membrane interactions and communication. J. Biol. Chem. 297, 101164 (2021).

    Article  Google Scholar 

  126. Upham, B. L. & Trosko, J. E. Oxidative-dependent integration of signal transduction with intercellular gap junctional communication in the control of gene expression. Antioxid. Redox Signal. 11, 297–307 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Bücher, T. et al. State of oxidation–reduction and state of binding in the cytosolic NADH-system as disclosed by equilibration with extracellular lactate-pyruvate in hemoglobin-free perfused rat liver. Eur. J. Biochem. 27, 301–317 (1972).

    Article  PubMed  Google Scholar 

  128. Hassinen, I. E. Signaling and regulation through the NAD(+) and NADP(+) networks. Antioxid. Redox Signal. 30, 857–874 (2019).

    Article  CAS  PubMed  Google Scholar 

  129. Lucaciu, S. A., Leighton, S. E., Hauser, A., Yee, R. & Laird, D. W. Diversity in connexin biology. J. Biol. Chem. 299, 105263 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Zhang, K. et al. The mutual interplay of redox signaling and connexins. J. Mol. Med. 99, 933–941 (2021).

    Article  PubMed  Google Scholar 

  131. van Niel, G. et al. Challenges and directions in studying cell–cell communication by extracellular vesicles. Nat. Rev. Mol. Cell Biol. 23, 369–382 (2022). This paper shows a comprehensive review on the role of extracellular vesicles in cell–cell communication.

    Article  PubMed  Google Scholar 

  132. Dixson, A. C., Dawson, T. R., Di Vizio, D. & Weaver, A. M. Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat. Rev. Mol. Cell Biol. 24, 454–476 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Zhang, W. J., Liu, R., Chen, Y. H., Wang, M. H. & Du, J. Crosstalk between oxidative stress and exosomes. Oxid. Med. Cell Longev. 2022, 3553617 (2022).

    PubMed  PubMed Central  Google Scholar 

  134. Hervera, A. et al. Reactive oxygen species regulate axonal regeneration through the release of exosomal NADPH oxidase 2 complexes into injured axons. Nat. Cell Biol. 20, 307–319 (2018).

    Article  CAS  PubMed  Google Scholar 

  135. Clarke-Bland, C. E., Bill, R. M. & Devitt, A. Emerging roles for AQP in mammalian extracellular vesicles. Biochim. Biophys. Acta Biomembr. 1864, 183826 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Fichman, Y., Rowland, L., Oliver, M. J. & Mittler, R. ROS are evolutionary conserved cell-to-cell stress signals. Proc. Natl Acad. Sci. USA 120, e2305496120 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Saraswathibhatla, A., Indana, D. & Chaudhuri, O. Cell–extracellular matrix mechanotransduction in 3D. Nat. Rev. Mol. Cell Biol. 24, 495–516 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Taniguchi, N. et al. Glyco-redox, a link between oxidative stress and changes of glycans: lessons from research on glutathione, reactive oxygen and nitrogen species to glycobiology. Arch. Biochem. Biophys. 595, 72–80 (2016).

    Article  CAS  PubMed  Google Scholar 

  139. Khoder-Agha, F. & Kietzmann, T. The glyco-redox interplay: principles and consequences on the role of reactive oxygen species during protein glycosylation. Redox Biol. 42, 101888 (2021). This paper shows a comprehensive overview of the relation of protein glycosylation to redox regulation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Chen, P. H., Chi, J. T. & Boyce, M. Functional crosstalk among oxidative stress and O-GlcNAc signaling pathways. Glycobiology 28, 556–564 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Tanaka, L. Y., Oliveira, P. V. S. & Laurindo, F. R. M. Peri/epicellular thiol oxidoreductases as mediators of extracellular redox signaling. Antioxid. Redox Signal. 33, 280–307 (2020). This work describes the role of extracellular thiol oxidoreductases in redox signalling.

    Article  CAS  PubMed  Google Scholar 

  142. Lorenzen, I., Eble, J. A. & Hanschmann, E. M. Thiol switches in membrane proteins — extracellular redox regulation in cell biology. Biol. Chem. 402, 253–269 (2021).

    Article  CAS  PubMed  Google Scholar 

  143. Palacio, P. L., Godoy, J. R., Aktas, O. & Hanschmann, E. M. Changing perspectives from oxidative stress to redox signaling-extracellular redox control in translational medicine. Antioxidants 11, 1181 (2022).

    Article  Google Scholar 

  144. Forman, H. J., Bernardo, A. & Davies, K. J. What is the concentration of hydrogen peroxide in blood and plasma? Arch. Biochem. Biophys. 603, 48–53 (2016).

    Article  CAS  PubMed  Google Scholar 

  145. Hosogi, S. et al. Plasma membrane anchored nanosensor for quantifying endogenous production of H(2)O(2) in living cells. Biosens. Bioelectron. 179, 113077 (2021).

    Article  CAS  PubMed  Google Scholar 

  146. Go, Y. M. & Jones, D. P. The redox proteome. J. Biol. Chem. 288, 26512–26520 (2013). This paper provides an overview of the fundamentals of the redox proteome.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Hawkins, C. L. & Davies, M. J. Detection, identification, and quantification of oxidative protein modifications. J. Biol. Chem. 294, 19683–19708 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Cortese-Krott, M. M. et al. The reactive species interactome: evolutionary emergence, biological significance, and opportunities for redox metabolomics and personalized medicine. Antioxid. Redox Signal. 27, 684–712 (2017). This paper describes how reactive species interact among themselves and with their targets.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Malard, E., Valable, S., Bernaudin, M., Pérès, E. & Chatre, L. The reactive species interactome in the brain. Antioxid. Redox Signal. 35, 1176–1206 (2021).

    Article  CAS  PubMed  Google Scholar 

  150. Go, Y. M., Chandler, J. D. & Jones, D. P. The cysteine proteome. Free Radic. Biol. Med. 84, 227–245 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Reczek, C. R. & Chandel, N. S. ROS-dependent signal transduction. Curr. Opin. Cell Biol. 33, 8–13 (2015).

    Article  CAS  PubMed  Google Scholar 

  152. Cadenas-Garrido, P. et al. Using redox proteomics to gain new insights into neurodegenerative disease and protein modification. Antioxidants 13, 127 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Xiao, H. et al. A quantitative tissue-specific landscape of protein redox regulation during aging. Cell 180, 968–983 (2020). This work describes ‘Oximouse’, a quantitative data set of the mouse cysteine redox proteome in vivo.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Pham, T. K., Buczek, W. A., Mead, R. J., Shaw, P. J. & Collins, M. O. Proteomic approaches to study cysteine oxidation: applications in neurodegenerative diseases. Front. Mol. Neurosci. 14, 678837 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Fu, L. et al. Nucleophilic covalent ligand discovery for the cysteine redoxome. Nat. Chem. Biol. 19, 1309–1319 (2023).

    Article  CAS  PubMed  Google Scholar 

  156. Ferreira, R. B., Fu, L., Jung, Y., Yang, J. & Carroll, K. S. Reaction-based fluorogenic probes for detecting protein cysteine oxidation in living cells. Nat. Commun. 13, 5522 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Erdogan, Y. C. et al. Complexities of the chemogenetic toolkit: differential mDAAO activation by D-amino substrates and subcellular targeting. Free Radic. Biol. Med. 177, 132–142 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Steinhorn, B. et al. Chemogenetic generation of hydrogen peroxide in the heart induces severe cardiac dysfunction. Nat. Commun. 9, 4044 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  159. Steinhorn, B., Eroglu, E. & Michel, T. Chemogenetic approaches to probe redox pathways: implications for cardiovascular pharmacology and toxicology. Annu. Rev. Pharmacol. Toxicol. 62, 551–571 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  160. Nanadikar, M. S. et al. IDH3γ functions as a redox switch regulating mitochondrial energy metabolism and contractility in the heart. Nat. Commun. 14, 2123 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Kritsiligkou, P. et al. Proteome-wide tagging with an H(2)O(2) biosensor reveals highly localized and dynamic redox microenvironments. Proc. Natl Acad. Sci. USA 120, e2314043120 (2023). This work describes the complex nature of cellular redox microenvironments.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Feng, S. et al. Development of a clickable probe for profiling of protein glutathionylation in the central cellular metabolism of E. coli and Drosophila. Chem. Biol. 22, 1461–1469 (2015).

    Article  CAS  PubMed  Google Scholar 

  163. van der Reest, J., Lilla, S., Zheng, L., Zanivan, S. & Gottlieb, E. Proteome-wide analysis of cysteine oxidation reveals metabolic sensitivity to redox stress. Nat. Commun. 9, 1581 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  164. Maret, W. The redox biology of redox-inert zinc ions. Free Radic. Biol. Med. 134, 311–326 (2019).

    Article  CAS  PubMed  Google Scholar 

  165. Hübner, C. & Haase, H. Interactions of zinc- and redox-signaling pathways. Redox Biol. 41, 101916 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  166. Netto, L. E. S. & Machado, L. Preferential redox regulation of cysteine-based protein tyrosine phosphatases: structural and biochemical diversity. FEBS J. 289, 5480–5504 (2022).

    Article  CAS  PubMed  Google Scholar 

  167. Tossounian, M. A., Zhang, B. & Gout, I. The writers, readers, and erasers in redox regulation of GAPDH. Antioxidants 9, 1288 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Mailloux, R. J. Protein S-glutathionylation reactions as a global inhibitor of cell metabolism for the desensitization of hydrogen peroxide signals. Redox Biol. 32, 101472 (2020). This paper examines the role of protein S-glutathionylation in redox signalling.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Bodnar, Y. & Lillig, C. H. Cysteinyl and methionyl redox switches: structural prerequisites and consequences. Redox Biol. 65, 102832 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Reichmann, D. & Jakob, U. The roles of conditional disorder in redox proteins. Curr. Opin. Struct. Biol. 23, 436–442 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Devant, P. et al. Gasdermin D pore-forming activity is redox-sensitive. Cell Rep. 42, 112008 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Weismiller, H. A., Holub, T. J., Krzesinski, B. J. & Margittai, M. A thiol-based intramolecular redox switch in four-repeat tau controls fibril assembly and disassembly. J. Biol. Chem. 297, 101021 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Ulrich, K. et al. From guide to guard-activation mechanism of the stress-sensing chaperone Get3. Mol. Cell 82, 3226–3238.e7 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Moskovitz, J. & Smith, A. Methionine sulfoxide and the methionine sulfoxide reductase system as modulators of signal transduction pathways: a review. Amino Acids 53, 1011–1020 (2021).

    Article  CAS  PubMed  Google Scholar 

  175. Lim, J. M., Kim, G. & Levine, R. L. Methionine in proteins: it’s not just for protein initiation anymore. Neurochem. Res. 44, 247–257 (2019).

    Article  CAS  PubMed  Google Scholar 

  176. Kaya, A., Lee, B. C. & Gladyshev, V. N. Regulation of protein function by reversible methionine oxidation and the role of selenoprotein MsrB1. Antioxid. Redox Signal. 23, 814–822 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Labunskyy, V. M., Hatfield, D. L. & Gladyshev, V. N. Selenoproteins: molecular pathways and physiological roles. Physiol. Rev. 94, 739–777 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Zhang, Y. et al. Role of selenoproteins in redox regulation of signaling and the antioxidant system: a review. Antioxidants (Basel) 9, 383 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Nasim, M. J., Zuraik, M. M., Abdin, A. Y., Ney, Y. & Jacob, C. Selenomethionine: a pink trojan redox horse with implications in aging and various age-related diseases. Antioxidants 10, 882 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Assmann, A., Briviba, K. & Sies, H. Reduction of methionine selenoxide to selenomethionine by glutathione. Arch. Biochem. Biophys. 349, 201–203 (1998).

    Article  CAS  PubMed  Google Scholar 

  181. Giulivi, C., Traaseth, N. J. & Davies, K. J. Tyrosine oxidation products: analysis and biological relevance. Amino Acids 25, 227–232 (2003).

    Article  CAS  PubMed  Google Scholar 

  182. Zhou, X., Liu, F., Li, N. & Zhang, Y. Large-scale qualitative and quantitative assessment of dityrosine crosslinking omics in response to endogenous and exogenous hydrogen peroxide in Escherichia coli. Antioxidants 12, 786 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Piacenza, L., Zeida, A., Trujillo, M. & Radi, R. The superoxide radical switch in the biology of nitric oxide and peroxynitrite. Physiol. Rev. 102, 1881–1906 (2022). This paper is a comprehensive review on the role of reactive nitrogen species in redox regulation.

    Article  CAS  PubMed  Google Scholar 

  184. Griswold-Prenner, I. et al. Unveiling the human nitroproteome: protein tyrosine nitration in cell signaling and cancer. J. Biol. Chem. 299, 105038 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Cobley, J. N. Oxiforms: unique cysteine residue- and chemotype-specified chemical combinations can produce functionally-distinct proteoforms. Bioessays 45, e2200248 (2023).

    Article  PubMed  Google Scholar 

  186. Cobley, J. N. 50 Shades of oxidative stress: a state-specific cysteine redox pattern hypothesis. Redox Biol. 67, 102936 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Pak, V. V. et al. Ultrasensitive genetically encoded indicator for hydrogen peroxide identifies roles for the oxidant in cell migration and mitochondrial function. Cell Metab. 31, 642–653 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Morgan, B. et al. Real-time monitoring of basal H2O2 levels with peroxiredoxin-based probes. Nat. Chem. Biol. 12, 437–443 (2016).

    Article  CAS  PubMed  Google Scholar 

  189. Radzinski, M. et al. Temporal profiling of redox-dependent heterogeneity in single cells. eLife 7, e37623 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  190. Braeckman, B. P., Smolders, A., Back, P. & De Henau, S. In vivo detection of reactive oxygen species and redox status in Caenorhabditis elegans. Antioxid. Redox Signal. 25, 577–592 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. 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 

  192. Bazopoulou, D. et al. Developmental ROS individualizes organismal stress resistance and lifespan. Nature 576, 301–305 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Janke, R., Dodson, A. E. & Rine, J. Metabolism and epigenetics. Annu. Rev. Cell Dev. Biol. 31, 473–496 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Parkhitko, A. A., Filine, E., Mohr, S. E., Moskalev, A. & Perrimon, N. Targeting metabolic pathways for extension of lifespan and healthspan across multiple species. Ageing Res. Rev. 64, 101188 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Oleson, B. J. et al. Early life changes in histone landscape protect against age-associated amyloid toxicities through HSF-1-dependent regulation of lipid metabolism. Nat. Aging 4, 48–61 (2024). This paper examines redox-regulated histone methylation in relation to the regulation of lipid metabolism.

    Article  CAS  PubMed  Google Scholar 

  196. Dustin, C. M., Heppner, D. E., Lin, M. J. & van der Vliet, A. Redox regulation of tyrosine kinase signalling: more than meets the eye. J. Biochem. 167, 151–163 (2020).

    Article  CAS  PubMed  Google Scholar 

  197. Mitchell, S., Vargas, J. & Hoffmann, A. Signaling via the NFκB system. Wiley Interdiscip. Rev. Syst. Biol. Med. 8, 227–241 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Truong, T. H. et al. Molecular basis for redox activation of epidermal growth factor receptor kinase. Cell Chem. Biol. 23, 837–848 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Benhar, M. Oxidants, antioxidants and thiol redox switches in the control of regulated cell death pathways. Antioxidants 9, 309 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Korshavn, K. J., Wales, T. E., Bird, G. H., Engen, J. R. & Walensky, L. D. A redox switch regulates the structure and function of anti-apoptotic BFL-1. Nat. Struct. Mol. Biol. 27, 781–789 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Braunstein, I. et al. Opposing effects of polysulfides and thioredoxin on apoptosis through caspase persulfidation. J. Biol. Chem. 295, 3590–3600 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Jiang, X., Stockwell, B. R. & Conrad, M. Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 22, 266–282 (2021). This study shows a comprehensive overview of the role of ferroptosis in redox biology.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Seiler, A. et al. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15-lipoxygenase dependent- and AIF-mediated cell death. Cell Metab. 8, 237–248 (2008).

    Article  CAS  PubMed  Google Scholar 

  204. Llabani, E. et al. Diverse compounds from pleuromutilin lead to a thioredoxin inhibitor and inducer of ferroptosis. Nat. Chem. 11, 521–532 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Noguchi, T. et al. Nuclear-accumulated SQSTM1/p62-based ALIS act as microdomains sensing cellular stresses and triggering oxidative stress-induced parthanatos. Cell Death Dis. 9, 1193 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Scaturro, P. & Pichlmair, A. Oxeiptosis: a discreet way to respond to radicals. Curr. Opin. Immunol. 56, 37–43 (2019).

    Article  CAS  PubMed  Google Scholar 

  207. Pagliaro, P. & Penna, C. Inhibitors of NLRP3 inflammasome in ischemic heart disease: focus on functional and redox aspects. Antioxidants 12, 1396 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Peters, A., Nawrot, T. S. & Baccarelli, A. A. Hallmarks of environmental insults. Cell 184, 1455–1468 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Larigot, L. et al. Aryl hydrocarbon receptor and its diverse ligands and functions: an exposome receptor. Annu. Rev. Pharmacol. Toxicol. 62, 383–404 (2022).

    Article  CAS  PubMed  Google Scholar 

  210. Go, Y. M. & Jones, D. P. Redox biology: interface of the exposome with the proteome, epigenome and genome. Redox Biol. 2, 358–360 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Salminen, A. Aryl hydrocarbon receptor (AhR) reveals evidence of antagonistic pleiotropy in the regulation of the aging process. Cell Mol. Life Sci. 79, 489 (2022). This study is an overview of the pleiotropic role of aryl hydrocarbon receptor in biology, with particular reference to ageing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Sondermann, N. C. et al. Functions of the aryl hydrocarbon receptor (AHR) beyond the canonical AHR/ARNT signaling pathway. Biochem. Pharmacol. 208, 115371 (2023).

    Article  CAS  PubMed  Google Scholar 

  213. Muri, J. & Kopf, M. Redox regulation of immunometabolism. Nat. Rev. Immunol. 21, 363–381 (2021). This review, together with that by Morris et al. (2022), summarizes the knowledge on redox regulation of the immune response.

    Article  CAS  PubMed  Google Scholar 

  214. Morris, G., Gevezova, M., Sarafian, V. & Maes, M. Redox regulation of the immune response. Cell Mol. Immunol. 19, 1079–1101 (2022). This review, together with that by Muri and Kopf (2021), summarizes the knowledge on redox regulation of the immune response.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Geng, S., Lin, R., Wu, Y., Wang, J. & Li, L. Modulation of innate immune memory dynamics by subcellular reactive oxygen species. Antioxid. Redox Signal. 39, 1027–1038 (2023).

    Article  CAS  PubMed  Google Scholar 

  216. Granados, J. C. et al. AHR is a master regulator of diverse pathways in endogenous metabolism. Sci. Rep. 12, 16625 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  217. Rius-Pérez, S., Torres-Cuevas, I., Millán, I., Ortega, Á. L. & Pérez, S. PGC-1α, inflammation, and oxidative stress: an integrative view in metabolism. Oxid. Med. Cell Longev. 2020, 1452696 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  218. Lui, P. Y., Jin, D. Y. & Stevenson, N. J. MicroRNA: master controllers of intracellular signaling pathways. Cell Mol. Life Sci. 72, 3531–3542 (2015).

    Article  CAS  PubMed  Google Scholar 

  219. Buler, M., Andersson, U. & Hakkola, J. Who watches the watchmen? Regulation of the expression and activity of sirtuins. FASEB J. 30, 3942–3960 (2016).

    Article  CAS  PubMed  Google Scholar 

  220. Putker, M. & O’Neill, J. S. Reciprocal control of the circadian clock and cellular redox state — a critical appraisal. Mol. Cell 39, 6–19 (2016).

    Article  CAS  Google Scholar 

  221. Di Mascio, P. et al. Singlet molecular oxygen reactions with nucleic acids, lipids, and proteins. Chem. Rev. 119, 2043–2086 (2019).

    Article  PubMed  Google Scholar 

  222. Brash, D. E. & Goncalves, L. C. P. Chemiexcitation: mammalian photochemistry in the dark. Photochem. Photobiol. 99, 251–276 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Spickett, C. M. Formation of oxidatively modified lipids as the basis for a cellular epilipidome. Front. Endocrinol. 11, 602771 (2020).

    Article  Google Scholar 

  224. Doka, E. et al. Control of protein function through oxidation and reduction of persulfidated states. Sci. Adv. 6, eaax8358 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Lopez-Otin, C. & Kroemer, G. Hallmarks of health. Cell 184, 33–63 (2021).

    Article  CAS  PubMed  Google Scholar 

  226. Ayres, J. S. The biology of physiological health. Cell 181, 250–269 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Lopez-Otin, C., Pietrocola, F., Roiz-Valle, D., Galluzzi, L. & Kroemer, G. Meta-hallmarks of aging and cancer. Cell Metab. 35, 12–35 (2023).

    Article  CAS  PubMed  Google Scholar 

  228. Feelisch, M., Cortese-Krott, M. M., Santolini, J., Wootton, S. A. & Jackson, A. A. Systems redox biology in health and disease. EXCLI J. 21, 623–646 (2022).

    PubMed  PubMed Central  Google Scholar 

  229. Heusch, G. et al. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol. 67, 102894 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Sörensen, M. et al. Health position paper and redox perspectives — disease burden by transportation noise. Redox Biol. 69, 102995 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  231. Meng, J. et al. Precision redox: the key for antioxidant pharmacology. Antioxid. Redox Signal. 34, 1069–1082 (2021). This paper describes the challenges for the future of precision redox-based therapy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Nogales, C. et al. Network pharmacology: curing causal mechanisms instead of treating symptoms. Trends Pharmacol. Sci. 43, 136–150 (2022).

    Article  CAS  PubMed  Google Scholar 

  233. Wang, R. S., Maron, B. A. & Loscalzo, J. Multiomics network medicine approaches to precision medicine and therapeutics in cardiovascular diseases. Arterioscler. Thromb. Vasc. Biol. 43, 493–503 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Halliwell, B. Understanding mechanisms of antioxidant action in health and disease. Nat. Rev. Mol. Cell Biol. 25, 13–33 (2024). This study is a comprehensive overview of mechanisms of antioxidant action.

    Article  CAS  PubMed  Google Scholar 

  235. Hayes, J. D., Dinkova-Kostova, A. T. & Tew, K. D. Oxidative stress in cancer. Cancer Cell 38, 167–197 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Zhang, J. et al. Systematic identification of anticancer drug targets reveals a nucleus-to-mitochondria ROS-sensing pathway. Cell 186, 2361–2379.e25 (2023).

    Article  CAS  PubMed  Google Scholar 

  237. Krafczyk, N. & Klotz, L. O. FOXO transcription factors in antioxidant defense. IUBMB Life 74, 53–61 (2022).

    Article  CAS  PubMed  Google Scholar 

  238. Rodriguez-Colman, M. J., Dansen, T. B. & Burgering, B. M. T. FOXO transcription factors as mediators of stress adaptation. Nat. Rev. Mol. Cell Biol. 25, 46–64 (2024).

    Article  CAS  PubMed  Google Scholar 

  239. Lee, P., Chandel, N. S. & Simon, M. C. Cellular adaptation to hypoxia through hypoxia inducible factors and beyond. Nat. Rev. Mol. Cell Biol. 21, 268–283 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Liu, X., Xu, C., Xiao, W. & Yan, N. Unravelling the role of NFE2L1 in stress responses and related diseases. Redox Biol. 65, 102819 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Katsuoka, F., Otsuki, A., Hatanaka, N., Okuyama, H. & Yamamoto, M. Target gene diversity of the Nrf1-MafG transcription factor revealed by a tethered heterodimer. Mol. Cell Biol. 42, e0052021 (2022).

    Article  PubMed  Google Scholar 

  242. Hu, S. et al. Nrf1 is an indispensable redox-determining factor for mitochondrial homeostasis by integrating multi-hierarchical regulatory networks. Redox Biol. 57, 102470 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Suzuki, T., Takahashi, J. & Yamamoto, M. Molecular basis of the KEAP1-NRF2 signaling pathway. Mol. Cell 46, 133–141 (2023).

    Article  CAS  Google Scholar 

  244. Ibrahim, L. et al. Defining the functional targets of cap‘n’collar transcription factors NRF1, NRF2, and NRF3. Antioxidants 9, 1025 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Choi, H. I. et al. PGC-1alpha attenuates hydrogen peroxide-induced apoptotic cell death by upregulating Nrf-2 via GSK3beta inactivation mediated by activated p38 in HK-2 cells. Sci. Rep. 7, 4319 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  246. Shi, T., van Soest, D. M. K., Polderman, P. E., Burgering, B. M. T. & Dansen, T. B. DNA damage and oxidant stress activate p53 through differential upstream signaling pathways. Free Radic. Biol. Med. 172, 298–311 (2021).

    Article  CAS  PubMed  Google Scholar 

  247. Rothhammer, V. & Quintana, F. J. The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat. Rev. Immunol. 19, 184–197 (2019).

    Article  CAS  PubMed  Google Scholar 

  248. Scholtes, C. & Giguère, V. Transcriptional regulation of ROS homeostasis by the ERR subfamily of nuclear receptors. Antioxidants 10, 437 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Truong, T. H. & Carroll, K. S. Redox regulation of epidermal growth factor receptor signaling through cysteine oxidation. Biochemistry 51, 9954–9965 (2012).

    Article  CAS  PubMed  Google Scholar 

  250. Pastore, A. & Piemonte, F. S-glutathionylation signaling in cell biology: progress and prospects. Eur. J. Pharm. Sci. 46, 279–292 (2012).

    Article  CAS  PubMed  Google Scholar 

  251. Carter, E. L. & Ragsdale, S. W. Modulation of nuclear receptor function by cellular redox poise. J. Inorg. Biochem. 133, 92–103 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Shao, D. et al. A redox-dependent mechanism for regulation of AMPK activation by thioredoxin1 during energy starvation. Cell Metab. 19, 232–245 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Zhao, Y. et al. ROS signaling under metabolic stress: cross-talk between AMPK and AKT pathway. Mol. Cancer 16, 79 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  254. Xiong, Y., Uys, J. D., Tew, K. D. & Townsend, D. M. S-glutathionylation: from molecular mechanisms to health outcomes. Antioxid. Redox Signal. 15, 233–270 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. Singh, C. K. et al. The role of sirtuins in antioxidant and redox signaling. Antioxid. Redox Signal. 28, 643–661 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Kalous, K. S., Wynia-Smith, S. L. & Smith, B. C. Sirtuin oxidative post-translational modifications. Front. Physiol. 12, 763417 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  257. Heppner, D. E. Structural insights into redox-active cysteine residues of the Src family kinases. Redox Biol. 41, 101934 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Wu, Y., Lim, Y. W. & Parton, R. G. Caveolae and the oxidative stress response. Biochem. Soc. Trans. 51, 1377–1385 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Monico, A., Guzman-Caldentey, J., Pajares, M. A., Martin-Santamaria, S. & Perez-Sala, D. Molecular insight into the regulation of vimentin by cysteine modifications and zinc binding. Antioxidants 10, 1039 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Zhang, Y. et al. RIP1 autophosphorylation is promoted by mitochondrial ROS and is essential for RIP3 recruitment into necrosome. Nat. Commun. 8, 14329 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  261. Mittler, R. & Jones, D. P. The redox code of plants. Plant Cell Environ. https://doi.org/10.1111/pce.14787 (2023).

  262. Oka, S. I., Titus, A. S., Zablocki, D. & Sadoshima, J. Molecular properties and regulation of NAD(+) kinase (NADK). Redox Biol. 59, 102561 (2023).

    Article  CAS  PubMed  Google Scholar 

  263. Kampjut, D. & Sazanov, L. A. Structure and mechanism of mitochondrial proton-translocating transhydrogenase. Nature 573, 291–295 (2019).

    Article  CAS  PubMed  Google Scholar 

  264. Francisco, A., Figueira, T. R. & Castilho, R. F. Mitochondrial NAD(P)(+) transhydrogenase: from molecular features to physiology and disease. Antioxid. Redox Signal. 36, 864–884 (2022).

    Article  CAS  PubMed  Google Scholar 

  265. Katsyuba, E., Romani, M., Hofer, D. & Auwerx, J. NAD(+) homeostasis in health and disease. Nat. Metab. 2, 9–31 (2020).

    Article  CAS  PubMed  Google Scholar 

  266. Netto, L. E. & Antunes, F. The roles of peroxiredoxin and thioredoxin in hydrogen peroxide sensing and in signal transduction. Mol. Cell 39, 65–71 (2016).

    Article  CAS  Google Scholar 

  267. Peskin, A. V. et al. The high reactivity of peroxiredoxin 2 with H(2)O(2) is not reflected in its reaction with other oxidants and thiol reagents. J. Biol. Chem. 282, 11885–11892 (2007).

    Article  CAS  PubMed  Google Scholar 

  268. Sobotta, M. C. et al. Peroxiredoxin-2 and STAT3 form a redox relay for H2O2 signaling. Nat. Chem. Biol. 11, 64–70 (2015). This study shows a prototypical example of redox relay in hydrogen peroxide signalling.

    Article  CAS  PubMed  Google Scholar 

  269. Garrido Ruiz, D., Sandoval-Perez, A., Rangarajan, A. V., Gunderson, E. L. & Jacobson, M. P. Cysteine oxidation in proteins: structure, biophysics, and simulation. Biochemistry 61, 2165–2176 (2022).

    Article  CAS  PubMed  Google Scholar 

  270. Chen, S. M. & Tang, X. Q. Homocysteinylation and sulfhydration in diseases. Curr. Neuropharmacol. 20, 1726–1735 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Gout, I. Coenzyme A, protein CoAlation and redox regulation in mammalian cells. Biochem. Soc. Trans. 46, 721–728 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. Nakamura, T., Oh, C. K., Zhang, X., Tannenbaum, S. R. & Lipton, S. A. Protein transnitrosylation signaling networks contribute to inflammaging and neurodegenerative disorders. Antioxid. Redox Signal. 35, 531–550 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. Mustafa, A. K. et al. H2S signals through protein S-sulfhydration. Sci. Signal. 2, ra72 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  274. Wensien, M. et al. A lysine–cysteine redox switch with an NOS bridge regulates enzyme function. Nature 593, 460–464 (2021).

    Article  CAS  PubMed  Google Scholar 

  275. Rabe von Pappenheim, F. et al. Widespread occurrence of covalent lysine–cysteine redox switches in proteins. Nat. Chem. Biol. 18, 368–375 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  276. Dennis, K. K., Go, Y. M. & Jones, D. P. Redox systems biology of nutrition and oxidative stress. J. Nutr. 149, 553–565 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  277. Vogel, C. F. A., Van Winkle, L. S., Esser, C. & Haarmann-Stemmann, T. The aryl hydrocarbon receptor as a target of environmental stressors — implications for pollution mediated stress and inflammatory responses. Redox Biol. 34, 101530 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  278. Gargaro, M. et al. The landscape of AhR regulators and coregulators to fine-tune AhR functions. Int. J. Mol. Sci. 22, 757 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Vogeley, C. et al. Unraveling the differential impact of PAHs and dioxin-like compounds on AKR1C3 reveals the EGFR extracellular domain as a critical determinant of the AHR response. Environ. Int. 158, 106989 (2022).

    Article  CAS  PubMed  Google Scholar 

  280. Hu, X. et al. A scalable workflow to characterize the human exposome. Nat. Commun. 12, 5575 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  281. Frenis, K. et al. Redox switches in noise-induced cardiovascular and neuronal dysregulation. Front. Mol. Biosci. 8, 784910 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  282. Münzel, T., Sorensen, M., Hahad, O., Nieuwenhuijsen, M. & Daiber, A. The contribution of the exposome to the burden of cardiovascular disease. Nat. Rev. Cardiol. 20, 651–669 (2023).

    Article  PubMed  Google Scholar 

  283. Krutmann, J., Schikowski, T., Morita, A. & Berneburg, M. Environmentally-induced (extrinsic) skin aging: exposomal factors and underlying mechanisms. J. Invest. Dermatol. 141, 1096–1103 (2021).

    Article  CAS  PubMed  Google Scholar 

  284. Ghezzi, P. & Rubartelli, A. Redox regulation of defense against bacterial and viral pathogens. Curr. Opin. Chem. Biol. 76, 102339 (2023).

    Article  CAS  PubMed  Google Scholar 

  285. Dumas, A. & Knaus, U. G. Raising the ‘good’ oxidants for immune protection. Front. Immunol. 12, 698042 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  286. Kunst, C. et al. The influence of gut microbiota on oxidative stress and the immune system. Biomedicines 11, 1388 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  287. Cobley, J. N. How exercise induces oxidative eustress. in Oxidative Stress: Eustress and Distress (ed. Sies, H.) 447–462 (Academic Press, 2020).

  288. Margaritelis, N. V., Paschalis, V., Theodorou, A. A., Kyparos, A. & Nikolaidis, M. G. Redox basis of exercise physiology. Redox Biol. 35, 101499 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  289. Lisi, V. et al. Steady-state redox status in circulating extracellular vesicles: a proof-of-principle study on the role of fitness level and short-term aerobic training in healthy young males. Free Radic. Biol. Med. 204, 266–275 (2023).

    Article  CAS  PubMed  Google Scholar 

  290. Felix-Soriano, E. & Stanford, K. I. Exerkines and redox homeostasis. Redox Biol. 63, 102748 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Radak, Z. et al. Epigenetic and ‘redoxogenetic’ adaptation to physical exercise. Free Radic. Biol. Med. 210, 65–74 (2024).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are grateful to D. P. Jones, a longtime friend and colleague, for his substantial input and advice. B. Oleson helped with preparing figures. The authors also thank many researchers active in redox biology, who have generated an impressive literature that could not be covered exhaustively in the present overview. H.S. acknowledges longstanding support by Deutsche Forschungsgemeinschaft, Bonn, Germany, and by the National Foundation for Cancer Research, Bethesda, MD, USA. R.J.M. acknowledges Natural Sciences and Engineering Research Council of Canada Discovery Grant Program (RGPIN-2022-03240). U.J. acknowledges the National Institute of Health grant R35 GM122506 and the National Institute of Aging grant R21 AG078540.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the Review.

Corresponding authors

Correspondence to Helmut Sies, Ryan J. Mailloux or Ursula Jakob.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Molecular Cell Biology thanks Tobias Dansen, who co-reviewed with Janneke Keijer, Peter Nagy and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

‘Oximouse’: https://oximouse.hms.harvard.edu

Glossary

Antioxidants

Any substance that prevents, delays or removes oxidative damage. In biology, this includes various antioxidant enzymes and low-molecular-weight antioxidant compounds.

Hormesis

This term describes a biphasic dose–response phenomenon, whereby low-dose exposure leads to stimulation or preconditioning and high-dose exposure causes inhibition. The phenomenon is often described by a J-shaped or inverted U-shaped dose–response curve.

Oxidative distress

Biomolecular damage upon supraphysiological exposure to oxidants, resulting in detrimental consequence to life processes.

p53 pathway

A tumour suppression pathway, which oversees cell cycle regulation, DNA repair and cell death mechanisms to halt the proliferation of aberrant cells.

Redox stress signalling threshold

(RST). Condition at which the oxidant or reductant load causes the transition from beneficial (eustress) to detrimental (distress) outcome.

Surface-enhanced Raman spectroscopy

(SERS). A surface-sensitive method that amplifies Raman scattering from molecules absorbed on metal surfaces or nanostructures.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sies, H., Mailloux, R.J. & Jakob, U. Fundamentals of redox regulation in biology. Nat Rev Mol Cell Biol (2024). https://doi.org/10.1038/s41580-024-00730-2

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41580-024-00730-2

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research