Skip to main content

Advertisement

Log in

Reduction–oxidation (redox) system in radiation-induced normal tissue injury: molecular mechanisms and implications in radiation therapeutics

  • Review Article
  • Published:
Clinical and Translational Oncology Aims and scope Submit manuscript

Abstract

Every year, millions of cancer patients undergo radiation therapy for treating and destroying abnormal cell growths within normal cell environmental conditions. Thus, ionizing radiation can have positive therapeutic effects on cancer cells as well as post-detrimental effects on surrounding normal tissues. Previous studies in the past years have proposed that the reduction and oxidation metabolism in cells changes in response to ionizing radiation and has a key role in radiation toxicity to normal tissue. Free radicals generated from ionizing radiation result in upregulation of cyclooxygenases (COXs), nitric oxide synthase (NOSs), lipoxygenases (LOXs) as well as nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase), and their effected changes in mitochondrial functions are markedly noticeable. Each of these enzymes is diversely expressed in multiple cells, tissues and organs in a specific manner. Overproduction of reactive oxygen radicals (ROS), reactive hydroxyl radical (ROH) and reactive nitrogen radicals (RNS) in multiple cellular environments in the affected nucleus, cell membranes, cytosol and mitochondria, and other organelles, can specifically affect the sensitive and modifying enzymes of the redox system and repair proteins that play a pivotal role in both early and late effects of radiation. In recent years, ionizing radiation has been known to affect the redox functions and metabolism of NADPH oxidases (NOXs) as well as having destabilizing and detrimental effects on directly and indirectly affected cells, tissues and organs. More noteworthy, chronic free radical production may continue for years, increasing the risk of carcinogenesis and other oxidative stress-driven degenerative diseases as well as pathologies, in addition to late effect complications of organ fibrosis. Hence, knowledge about the mechanisms of chronic oxidative damage and injury in affected cells, tissues and organs following exposure to ionizing radiation may help in the development of treatment and management strategies of complications associated with radiotherapy (RT) or radiation accident victims. Thus, this medically relevant phenomenon may lead to the discovery of potential antioxidants and inhibitors with promising results in targeting and modulating the ROS/NO-sensitive enzymes in irradiated tissues and organ injury systems.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67(1):7–30.

    Article  PubMed  Google Scholar 

  2. Mettler FA Jr, Voelz GL. Major radiation exposure—what to expect and how to respond. N Engl J Med. 2002;346(20):1554–61.

    Article  PubMed  Google Scholar 

  3. Ring JP. Radiation risks and dirty bombs. Health Phys. 2004;86(2 Suppl):S42–7.

    Article  PubMed  CAS  Google Scholar 

  4. Mothersill C, Seymour CB. Radiation-induced bystander effects and the DNA paradigm: an “out of field” perspective. Mutat Res. 2006;597(1–2):5–10.

    Article  PubMed  CAS  Google Scholar 

  5. Spitz DR, Azzam EI, Li JJ, Gius D. Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation: a unifying concept in stress response biology. Cancer Metastasis Rev. 2004;23(3–4):311–22.

    Article  PubMed  CAS  Google Scholar 

  6. Sieber F, Muir SA, Cohen EP, North PE, Fish BL, Irving AA, et al. High-dose selenium for the mitigation of radiation injury: a pilot study in a rat model. Radiat Res. 2009;171(3):368–73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Zhao W, Robbins ME. Inflammation and chronic oxidative stress in radiation-induced late normal tissue injury: therapeutic implications. Curr Med Chem. 2009;16(2):130–43.

    Article  PubMed  CAS  Google Scholar 

  8. Najafi M, Shirazi A, Motevaseli E, Geraily Gh, Norouzi F, Heidari M, Rezapoor S. The melatonin immunomodulatory actions in radiotherapy. Biophys Rev. 2017;9(2):139–48.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Bae YS, Oh H, Rhee SG, Yoo YD. Regulation of reactive oxygen species generation in cell signaling. Mol Cells. 2011;32(6):491–509.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Birben E, Sahiner UM, Sackesen C, Erzurum S, Kalayci O. Oxidative stress and antioxidant defense. World Allergy Organ J. 2012;5(1):9–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Najafi M, Fardid R, Hadadi G, Fardid M. The mechanisms of radiation-induced bystander effect. J Biomed Phys Eng. 2014;4(4):163–72.

    PubMed  PubMed Central  CAS  Google Scholar 

  12. Pugin J. How tissue injury alarms the immune system and causes a systemic inflammatory response syndrome. Ann Intensive Care. 2012;2:27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Frey B, Hehlgans S, Rodel F, Gaipl US. Modulation of inflammation by low and high doses of ionizing radiation: implications for benign and malign diseases. Cancer Lett. 2015;368(2):230–7.

    Article  PubMed  CAS  Google Scholar 

  14. Frey B, Ruckert M, Deloch L, Ruhle PF, Derer A, Fietkau R, et al. Immunomodulation by ionizing radiation-impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol Rev. 2017;280(1):231–48.

    Article  PubMed  CAS  Google Scholar 

  15. Yahyapour R, Amini P, Rezapoor S, Rezaeyan A, Farhood B, Cheki M, et al. Targeting of inflammation for radiation protection and mitigation. Curr Mol Pharmacol. 2018. https://doi.org/10.2174/1874467210666171108165641.

    Article  PubMed  Google Scholar 

  16. Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007;87(1):245–313.

    Article  PubMed  CAS  Google Scholar 

  17. Drummond GR, Selemidis S, Griendling KK, Sobey CG. Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets. Nat Rev Drug Discov. 2011;10(6):453–71.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol. 2004;4(3):181–9.

    Article  PubMed  CAS  Google Scholar 

  19. Panday A, Sahoo MK, Osorio D, Batra S. NADPH oxidases: an overview from structure to innate immunity-associated pathologies. Cell Mol Immunol. 2015;12(1):5–23.

    Article  PubMed  CAS  Google Scholar 

  20. Fu X-J, Peng Y-B, Hu Y-P, Shi Y-Z, Yao M, Zhang X. NADPH oxidase 1 and its derived reactive oxygen species mediated tissue injury and repair. Oxid Med Cell Longev. 2014;2014:282854. https://doi.org/10.1155/2014/282854.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Sun Z, Liu F. Association of Nox1 and vinculin with colon cancer progression. Cancer Invest. 2013;31(4):273–8.

    Article  PubMed  CAS  Google Scholar 

  22. Liu F, Garcia AMG, Meyskens FL. NADPH oxidase 1 overexpression enhances invasion via matrix metalloproteinase-2 and epithelial–mesenchymal transition in melanoma cells. J Investig Dermatol. 2012;132(8):2033–41.

    Article  PubMed  CAS  Google Scholar 

  23. Choi S-H, Kim M, Lee H-J, Kim E-H, Kim C-H, Lee Y-J. Effects of NOX1 on fibroblastic changes of endothelial cells in radiation-induced pulmonary fibrosis. Mol Med Rep. 2016;13(5):4135–42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Jendrysik MA, Vasilevsky S, Yi L, Wood A, Zhu N, Zhao Y, et al. NADPH oxidase-2 derived ROS dictates murine DC cytokine-mediated cell fate decisions during CD4 T helper-cell commitment. PLoS ONE. 2011;6(12):e28198.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Narayanan P, Goodwin EH, Lehnert B. α particles initiate biological production of superoxide anions and hydrogen peroxide in human cells. Cancer Res. 1997;57(18):3963–71.

    PubMed  CAS  Google Scholar 

  26. Azzam EI, de Toledo SM, Spitz DR, Little JB. Oxidative metabolism modulates signal transduction and micronucleus formation in bystander cells from α-particle-irradiated normal human fibroblast cultures. Cancer Res. 2002;62(19):5436–42.

    PubMed  CAS  Google Scholar 

  27. Little J, Azzam E, De Toledo S, Nagasawa H. Bystander effects: intercellular transmission of radiation damage signals. Radiat Prot Dosim. 2002;99(1–4):159–62.

    Article  CAS  Google Scholar 

  28. Paillas S, Ladjohounlou R, Lozza C, Pichard A, Boudousq V, Jarlier M, et al. Localized irradiation of cell membrane by auger electrons is cytotoxic through oxidative stress-mediated nontargeted effects. Antioxid Redox Signal. 2016;25(8):467–84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Kim JH, Kim KM, Jung MH, Jung JH, Kang KM, Jeong BK, et al. Protective effects of alpha lipoic acid on radiation-induced salivary gland injury in rats. Oncotarget. 2016;7(20):29143–53.

    PubMed  PubMed Central  Google Scholar 

  30. Datta K, Suman S, Kallakury BV, Fornace AJ Jr. Exposure to heavy ion radiation induces persistent oxidative stress in mouse intestine. PLoS ONE. 2012;7(8):e42224.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Shin YS, Shin HA, Kang SU, Kim JH, Oh YT, Park KH, et al. Effect of epicatechin against radiation-induced oral mucositis: in vitro and in vivo study. PLoS ONE. 2013;8(7):e69151.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Pazhanisamy SK, Li H, Wang Y, Batinic-Haberle I, Zhou D. NADPH oxidase inhibition attenuates total body irradiation-induced haematopoietic genomic instability. Mutagenesis. 2011;26(3):431–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Wang Y, Liu L, Pazhanisamy SK, Li H, Meng A, Zhou D. Total body irradiation causes residual bone marrow injury by induction of persistent oxidative stress in murine hematopoietic stem cells. Free Radic Biol Med. 2010;48(2):348–56. https://doi.org/10.1016/j.freeradbiomed.2009.11.005.

    Article  PubMed  CAS  Google Scholar 

  34. Chang J, Feng W, Wang Y, Luo Y, Allen AR, Koturbash I, et al. Whole-body proton irradiation causes long-term damage to hematopoietic stem cells in mice. Radiat Res. 2015;183(2):240–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Zhang H, Y-a Wang, Meng A, Yan H, Wang X, Niu J, et al. Inhibiting TGFβ1 has a protective effect on mouse bone marrow suppression following ionizing radiation exposure in vitro. J Radiat Res. 2013;54(4):630–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Deng S, Yu K, Zhang B, Yao Y, Wang Z, Zhang J, et al. Toll-like receptor 4 promotes NO synthesis by upregulating GCHI expression under oxidative stress conditions in sheep monocytes/macrophages. Oxid Med Cell Longev. 2015;2015:359315. https://doi.org/10.1155/2015/359315.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Banfi B, Molnar G, Maturana A, Steger K, Hegedus B, Demaurex N, et al. A Ca(2+)-activated NADPH oxidase in testis, spleen, and lymph nodes. J Biol Chem. 2001;276(40):37594–601.

    Article  PubMed  CAS  Google Scholar 

  38. Weyemi U, Redon CE, Aziz T, Choudhuri R, Maeda D, Parekh PR, et al. Inactivation of NADPH oxidases NOX4 and NOX5 protects human primary fibroblasts from ionizing radiation-induced DNA damage. Radiat Res. 2015;183(3):262–70.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Ameziane-El-Hassani R, Talbot M, de Souza Dos Santos MC, Al Ghuzlan A, Hartl D, Bidart J-M, et al. NADPH oxidase DUOX1 promotes long-term persistence of oxidative stress after an exposure to irradiation. Proc Natl Acad Sci USA. 2015;112(16):5051–6.

    Article  PubMed  CAS  Google Scholar 

  40. Wu Y, Doroshow JH. Abstract 5358: IL-4/IL-13 induce Duox2/DuoxA2 expression and reactive oxygen production in human pancreatic and colon cancer cells. Cancer research. 2014;74(19 Supp):5358.

    Article  Google Scholar 

  41. Song J, Wei Y, Chen Q, Xing D. Cyclooxygenase 2-mediated apoptotic and inflammatory responses in photodynamic therapy treated breast adenocarcinoma cells and xenografts. J Photochem Photobiol B. 2014;134:27–36.

    Article  PubMed  CAS  Google Scholar 

  42. Steinauer KK, Gibbs I, Ning S, French JN, Armstrong J, Knox SJ. Radiation induces upregulation of cyclooxygenase-2 (COX-2) protein in PC-3 cells. Int J Radiat Oncol Biol Phys. 2000;48(2):325–8.

    Article  PubMed  CAS  Google Scholar 

  43. Wang D, DuBois RN. The role of COX-2 in intestinal inflammation and colorectal cancer. Oncogene. 2010;29(6):781–8.

    Article  PubMed  CAS  Google Scholar 

  44. Laube M, Kniess T, Pietzsch J. Development of antioxidant COX-2 inhibitors as radioprotective agents for radiation therapy—a hypothesis-driven review. Antioxidants (Basel). 2016;5(2):14.

    Article  PubMed Central  CAS  Google Scholar 

  45. Rezaeyan A, Haddadi GH, Hosseinzadeh M, Moradi M, Najafi M. Radioprotective effects of hesperidin on oxidative damages and histopathological changes induced by X-irradiation in rats heart tissue. J Med Phys. 2016;41(3):182–91.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Lee Y-K, Park SY, Kim Y-M, Lee WS, Park OJ. AMP kinase/cyclooxygenase-2 pathway regulates proliferation and apoptosis of cancer cells treated with quercetin. Exp Mol Med. 2009;41(3):201–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Sobolewski C, Cerella C, Dicato M, Ghibelli L, Diederich M. The role of cyclooxygenase-2 in cell proliferation and cell death in human malignancies. Int J Cell Biol. 2010;2010:215158.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Yahyapour R, Motevaseli E, Rezaeyan A, Abdollahi H, Farhood B, Cheki M, et al. Mechanisms of radiation bystander and non-targeted effects: implications to radiation carcinogenesis and radiotherapy. Curr Radiopharm. 2017. https://doi.org/10.2174/1874471011666171229123130.

    Article  Google Scholar 

  49. Chai Y, Calaf GM, Zhou H, Ghandhi SA, Elliston CD, Wen G, et al. Radiation induced COX-2 expression and mutagenesis at non-targeted lung tissues of gpt delta transgenic mice. Br J Cancer. 2013;108(1):91–8. https://doi.org/10.1038/bjc.2012.498.

    Article  PubMed  CAS  Google Scholar 

  50. Maccarrone M. Lipoxygenases, apoptosis, and the role of antioxidants. In: Demmig-Adams B, Adams WW, Mattoo AK, editors. Photoprotection, photoinhibition, gene regulation, and environment. Dordrecht: Springer; 2008. p. 321–32.

  51. Grichenko O, Shaposhnikova V, Levitman M, Kudriavtsev A, Korystov I. A study of the role of lipoxygenases in radiation-induced apoptosis of thymocytes. Inhibitory analysis. Radiatsionnaia biologiia, radioecologiia/Rossiiskaia akademiia nauk. 2003;44(1):27–31.

    Google Scholar 

  52. Matyshevskaia OP, Pastukh VN, Solodushko VA. Inhibition of lipoxygenase activity reduces radiation-induced DNA fragmentation in lymphocytes. Radiats Biol Radioecol. 1999;39(2–3):282–6.

    PubMed  CAS  Google Scholar 

  53. Grichenko OE, Pushin AC, Shaposhnikova VV, Levitman M, Korystov IuN. Analysis of 15-lipoxygenase activity in irradiated thymocytes. Izv Akad Nauk Ser Biol. 2004;5:517–21.

    Google Scholar 

  54. Aktan F. iNOS-mediated nitric oxide production and its regulation. Life Sci. 2004;75(6):639–53.

    Article  PubMed  CAS  Google Scholar 

  55. Ohta S, Matsuda S, Gunji M, Kamogawa A. The role of nitric oxide in radiation damage. Biol Pharm Bull. 2007;30(6):1102–7.

    Article  PubMed  CAS  Google Scholar 

  56. Kiang JG, Smith JT, Agravante NG. Geldanamycin analog 17-DMAG inhibits iNOS and caspases in gamma-irradiated human T cells. Radiat Res. 2009;172(3):321–30.

    Article  PubMed  CAS  Google Scholar 

  57. Malaviya R, Gow AJ, Francis M, Abramova EV, Laskin JD, Laskin DL. Radiation-induced lung injury and inflammation in mice: role of inducible nitric oxide synthase and surfactant protein D. Toxicol Sci. 2015;144(1):27–38.

    Article  PubMed  CAS  Google Scholar 

  58. Yakovlev VA. Role of nitric oxide in the radiation-induced bystander effect. Redox Biol. 2015;6:396–400.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Yakovlev VA. Nitric oxide dependent downregulation of BRCA1 expression promotes genetic instability. Cancer Res. 2013;73(2):706–15.

    Article  PubMed  CAS  Google Scholar 

  60. Giaid A, Lehnert SM, Chehayeb B, Chehayeb D, Kaplan I, Shenouda G. Inducible nitric oxide synthase and nitrotyrosine in mice with radiation-induced lung damage. Am J Clin Oncol. 2003;26(4):e67–72.

    PubMed  Google Scholar 

  61. Lahtz C, Pfeifer GP. Epigenetic changes of DNA repair genes in cancer. J Mol Cell Biol. 2011;3(1):51–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Kidane D, Chae WJ, Czochor J, Eckert KA, Glazer PM, Bothwell ALM, et al. Interplay between DNA repair and inflammation, and the link to cancer. Crit Rev Biochem Mol Biol. 2014;49(2):116–39.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Wink DA, Laval J. The Fpg protein, a DNA repair enzyme, is inhibited by the biomediator nitric oxide in vitro and in vivo. Carcinogenesis. 1994;15(10):2125–9.

    Article  PubMed  CAS  Google Scholar 

  64. Rezapoor S, Shirazi A, Abbasi S, Bazzaz JT, Izadi P, Rezaeejam H, et al. Modulation of radiation-induced base excision repair pathway gene expression by melatonin. J Med Phys. 2017;42(4):245–50.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Shinmura K, Kohno T, Kasai H, Koda K, Sugimura H, Yokota J. Infrequent mutations of the hOGG1 gene, that is involved in the excision of 8-hydroxyguanine in damaged DNA, in human gastric cancer. Cancer Sci. 1998;89(8):825–8.

    CAS  Google Scholar 

  66. Chevillard S, Radicella JP, Levalois C, Lebeau J, Poupon M-F, Oudard S, et al. Mutations in OGG1, a gene involved in the repair of oxidative DNA damage, are found in human lung and kidney tumours. Oncogene. 1998;16(23):3083–6.

    Article  PubMed  CAS  Google Scholar 

  67. Rolfe DF, Brown GC. Cellular energy utilization and molecular origin of standard metabolic rate in mammals. Physiol Rev. 1997;77(3):731–58.

    Article  PubMed  CAS  Google Scholar 

  68. Brand MD, Affourtit C, Esteves TC, Green K, Lambert AJ, Miwa S, et al. Mitochondrial superoxide: production, biological effects, and activation of uncoupling proteins. Free Radic Biol Med. 2004;37(6):755–67.

    Article  PubMed  CAS  Google Scholar 

  69. Tian L, Cai Q, Wei H. Alterations of antioxidant enzymes and oxidative damage to macromolecules in different organs of rats during aging. Free Radic Biol Med. 1998;24(9):1477–84.

    Article  PubMed  CAS  Google Scholar 

  70. Packer MA, Murphy MP. Peroxynitrite formed by simultaneous nitric oxide and superoxide generation causes cyclosporin-A-sensitive mitochondrial calcium efflux and depolarisation. Eur J Biochem. 1995;234(1):231–9.

    Article  PubMed  CAS  Google Scholar 

  71. Yamamori T, Yasui H, Yamazumi M, Wada Y, Nakamura Y, Nakamura H, et al. Ionizing radiation induces mitochondrial reactive oxygen species production accompanied by upregulation of mitochondrial electron transport chain function and mitochondrial content under control of the cell cycle checkpoint. Free Radic Biol Med. 2012;53(2):260–70.

    Article  PubMed  CAS  Google Scholar 

  72. Nugent SME, Mothersill CE, Seymour C, McClean B, Lyng FM, Murphy JEJ. Increased mitochondrial mass in cells with functionally compromised mitochondria after exposure to both direct γ radiation and bystander factors. Radiat Res. 2007;168(1):134–42.

    Article  PubMed  CAS  Google Scholar 

  73. Wang L, Kuwahara Y, Li L, Baba T, Shin R-W, Ohkubo Y, et al. Analysis of common deletion (CD) and a novel deletion of mitochondrial DNA induced by ionizing radiation. Int J Radiat Biol. 2007;83(7):433–42.

    Article  PubMed  CAS  Google Scholar 

  74. Tulard A, Hoffschir F, de Boisferon FH, Luccioni C, Bravard A. Persistent oxidative stress after ionizing radiation is involved in inherited radiosensitivity. Free Radic Biol Med. 2003;35(1):68–77.

    Article  PubMed  CAS  Google Scholar 

  75. Kobashigawa S, Suzuki K, Yamashita S. Ionizing radiation accelerates Drp1-dependent mitochondrial fission, which involves delayed mitochondrial reactive oxygen species production in normal human fibroblast-like cells. Biochem Biophys Res Commun. 2011;414(4):795–800.

    Article  PubMed  CAS  Google Scholar 

  76. Hosoki A, Yonekura S-I, Qing-Li Z, Zheng-Li W, Takasaki I, Tabuchi Y, et al. Mitochondria-targeted superoxide dismutase (SOD2) regulates radiation resistance and radiation stress response in HeLa cells. J Radiat Res. 2012;53(1):58–71.

    Article  PubMed  CAS  Google Scholar 

  77. Ogura A, Oowada S, Kon Y, Hirayama A, Yasui H, Meike S, et al. Redox regulation in radiation-induced cytochrome c release from mitochondria of human lung carcinoma A549 cells. Cancer Lett. 2009;277(1):64–71.

    Article  PubMed  CAS  Google Scholar 

  78. Yoshida T, Goto S, Kawakatsu M, Urata Y, Li T-S. Mitochondrial dysfunction, a probable cause of persistent oxidative stress after exposure to ionizing radiation. Free Radic Res. 2012;46(2):147–53.

    Article  PubMed  CAS  Google Scholar 

  79. Choi KM, Kang CM, Cho ES, Kang SM, Lee SB, Um HD. Ionizing radiation-induced micronucleus formation is mediated by reactive oxygen species that are produced in a manner dependent on mitochondria, Nox1, and JNK. Oncol Rep. 2007;17(5):1183–8.

    PubMed  CAS  Google Scholar 

  80. Copeland WC, Wachsman JT, Johnson FM, Penta JS. Mitochondrial DNA alterations in cancer. Cancer Invest. 2002;20:557–69.

    Article  PubMed  CAS  Google Scholar 

  81. Laurent A, Nicco C, Chereau C, et al. Controlling tumor growth by modulating endogenous production of reactive oxygen species. Cancer Res. 2005;65:948–56.

    PubMed  CAS  Google Scholar 

  82. Hajnoczky G, Csordas G, Das S, Garcia-Perez C, Saotome M, Sinha Roy S, et al. Mitochondrial calcium signalling and cell death: approaches for assessing the role of mitochondrial Ca2+ uptake in apoptosis. Cell Calcium. 2006;40(5–6):553–60.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Murphy MP. Mitochondrial dysfunction indirectly elevates ROS production by the endoplasmic reticulum. Cell Metab. 2013;18(2):145–6.

    Article  PubMed  CAS  Google Scholar 

  84. Barjaktarovic Z, Schmaltz D, Shyla A, Azimzadeh O, Schulz S, Haagen J, et al. Radiation-induced signaling results in mitochondrial impairment in mouse heart at 4 weeks after exposure to X-rays. PLoS ONE. 2011;6(12):e27811.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Kim GJ, Fiskum GM, Morgan WF. A role for mitochondrial dysfunction in perpetuating radiation-induced genomic instability. Cancer Res. 2006;66(21):10377–83.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Dikalov S. Cross talk between mitochondria and NADPH oxidases. Free Radic Biol Med. 2011;51(7):1289–301.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Ago T, Kuroda J, Pain J, Fu C, Li H, Sadoshima J. Upregulation of Nox4 by hypertrophic stimuli promotes apoptosis and mitochondrial dysfunction in cardiac myocytes. Circ Res. 2010;106(7):1253–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Kowluru RA, Mishra M. Oxidative stress, mitochondrial damage and diabetic retinopathy. Biochim Biophys Acta. 2015;1852(11):2474–83.

    Article  PubMed  CAS  Google Scholar 

  89. Afanas’ev I. Mechanisms of superoxide signaling in epigenetic processes: relation to aging and cancer. Aging Dis. 2015;6(3):216–27.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Simone NL, Soule BP, Ly D, Saleh AD, Savage JE, Degraff W, et al. Ionizing radiation-induced oxidative stress alters miRNA expression. PLoS ONE. 2009;4(7):e6377.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Ye L, Yu G, Wang C, Du B, Sun D, Liu J, et al. MicroRNA128a, BMI1 polycomb ring finger oncogene, and reactive oxygen species inhibit the growth of U87 MG glioblastoma cells following exposure to Xray radiation. Mol Med Rep. 2015;12(4):6247–54.

    Article  PubMed  CAS  Google Scholar 

  92. Chaudhry MA, Omaruddin RA, Brumbaugh CD, Tariq MA, Pourmand N. Identification of radiation-induced microRNA transcriptome by next-generation massively parallel sequencing. J Radiat Res. 2013;54(5):808–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Ling M, Li Y, Xu Y, Pang Y, Shen L, Jiang R, et al. Regulation of miRNA-21 by reactive oxygen species-activated ERK/NF-kappaB in arsenite-induced cell transformation. Free Radic Biol Med. 2012;52(9):1508–18.

    Article  PubMed  CAS  Google Scholar 

  94. Zhang X, Ng W-L, Wang P, Tian L, Werner E, Wang H, et al. MicroRNA-21 modulates the levels of reactive oxygen species levels by targeting SOD3 and TNFα. Can Res. 2012;72(18):4707–13.

    Article  CAS  Google Scholar 

  95. Xu S, Ding N, Pei H, Hu W, Wei W, Zhang X, et al. MiR-21 is involved in radiation-induced bystander effects. RNA Biol. 2014;11(9):1161–70.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Tian W, Yin X, Wang L, Wang J, Zhu W, Cao J, et al. The key role of miR-21-regulated SOD2 in the medium-mediated bystander responses in human fibroblasts induced by alpha-irradiated keratinocytes. Mutat Res. 2015;780:77–85.

    Article  PubMed  CAS  Google Scholar 

  97. Jiang Y, Chen X, Tian W, Yin X, Wang J, Yang H. The role of TGF-β1–miR-21–ROS pathway in bystander responses induced by irradiated non-small-cell lung cancer cells. Br J Cancer. 2014;111(4):772–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Rathore R, Zheng YM, Niu CF, Liu QH, Korde A, Ho YS, et al. Hypoxia activates NADPH oxidase to increase [ROS]i and [Ca2 +]i through the mitochondrial ROS-PKCepsilon signaling axis in pulmonary artery smooth muscle cells. Free Radic Biol Med. 2008;45(9):1223–31.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Marshall C, Mamary AJ, Verhoeven AJ, Marshall BE. Pulmonary artery NADPH-oxidase is activated in hypoxic pulmonary vasoconstriction. Am J Respir Cell Mol Biol. 1996;15(5):633–44.

    Article  PubMed  CAS  Google Scholar 

  100. Halberg N, Khan T, Trujillo ME, Wernstedt-Asterholm I, Attie AD, Sherwani S, et al. Hypoxia-inducible factor 1alpha induces fibrosis and insulin resistance in white adipose tissue. Mol Cell Biol. 2009;29(16):4467–83.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Vujaskovic Z, Anscher MS, Feng QF, Rabbani ZN, Amin K, Samulski TS, et al. Radiation-induced hypoxia may perpetuate late normal tissue injury. Int J Radiat Oncol Biol Phys. 2001;50(4):851–5.

    Article  PubMed  CAS  Google Scholar 

  102. Choi YJ, Kim HS, Lee J, Chung J, Lee JS, Choi JS, et al. Down-regulation of oxidative stress and COX-2 and iNOS expressions by dimethyl lithospermate in aged rat kidney. Arch Pharm Res. 2014;37(8):1032–8.

    Article  PubMed  CAS  Google Scholar 

  103. Hei TK, Zhou H, Ivanov VN, Hong M, Lieberman HB, Brenner DJ, et al. Mechanism of radiation-induced bystander effects: a unifying model. J Pharm Pharmacol. 2008;60(8):943–50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Edwards GO, Botchway SW, Hirst G, Wharton CW, Chipman JK, Meldrum RA. Gap junction communication dynamics and bystander effects from ultrasoft X-rays. Br J Cancer. 2004;90(7):1450–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Suzuki M, Tsuruoka C. Heavy charged particles produce a bystander effect via cell–cell junctions. Biol Sci Space. 2004;18(4):241–6.

    Article  PubMed  Google Scholar 

  106. Shao C, Furusawa Y, Aoki M, Ando K. Role of gap junctional intercellular communication in radiation-induced bystander effects in human fibroblasts. Radiat Res. 2003;160(3):318–23.

    Article  PubMed  CAS  Google Scholar 

  107. Bishayee A, Rao DV, Howell RW. Evidence for pronounced bystander effects caused by nonuniform distributions of radioactivity using a novel three-dimensional tissue culture model. Radiat Res. 1999;152(1):88–97.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Azzam EI, de Toledo SM, Little JB. Direct evidence for the participation of gap junction-mediated intercellular communication in the transmission of damage signals from α-particle irradiated to nonirradiated cells. Proc Natl Acad Sci USA. 2001;98(2):473–8.

    PubMed  CAS  Google Scholar 

  109. Najafi M, Shirazi A, Motevaseli E, Rezaeyan A, Salajegheh A, Rezapoor S. Melatonin as an anti-inflammatory agent in radiotherapy. Inflammopharmacology. 2017;25(4):403–13.

    Article  PubMed  CAS  Google Scholar 

  110. Morgan WF. Non-targeted and delayed effects of exposure to ionizing radiation: I. Radiation-induced genomic instability and bystander effects in vitro. 2003. Radiat Res. 2012;178(2):Av223–36.

    Article  PubMed  CAS  Google Scholar 

  111. Ghobadi A, Shirazi A, Najafi M, Kahkesh MH, Rezapoor S. Melatonin ameliorates radiation-induced oxidative stress at targeted and nontargeted lung tissue. J Med Phys. 2017;42(4):241.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Chai Y, Lam RK, Calaf GM, Zhou H, Amundson S, Hei TK. Radiation-induced non-targeted response in vivo: role of the TGFbeta-TGFBR1-COX-2 signalling pathway. Br J Cancer. 2013;108(5):1106–12.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Najafi MSA, Rezaeyan A. Bystander effect and second primary cancers following radiotherapy: what are its significances? J Med Phys. 2017;42:55–6.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Negrini S, Gorgoulis VG, Halazonetis TD. Genomic instability—an evolving hallmark of cancer. Nat Rev Mol Cell Biol. 2010;11(3):220–8.

    Article  PubMed  CAS  Google Scholar 

  115. Ferguson LR, Chen H, Collins AR, Connell M, Damia G, Dasgupta S, et al. Genomic instability in human cancer: Molecular insights and opportunities for therapeutic attack and prevention through diet and nutrition. Semin cancer biol. 2015;35(Suppl):S5–24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Goel A, Nagasaka T, Arnold CN, Inoue T, Hamilton C, Niedzwiecki D, et al. The CpG island methylator phenotype and chromosomal instability are inversely correlated in sporadic colorectal cancer. Gastroenterology. 2007;132(1):127–38.

    Article  PubMed  CAS  Google Scholar 

  117. Chatterjee A, Dasgupta S, Sidransky D. Mitochondrial subversion in cancer. Cancer Prev Res. 2011;4(5):638–54.

    Article  CAS  Google Scholar 

  118. Cook CC, Kim A, Terao S, Gotoh A, Higuchi M. Consumption of oxygen: a mitochondrial-generated progression signal of advanced cancer. Cell Death Dis. 2012;3(1):e258.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Konki M, Pasumarthy K, Malonzo M, Sainio A, Valensisi C, Söderström M, et al. Epigenetic silencing of the key antioxidant enzyme catalase in karyotypically abnormal human pluripotent stem cells. Sci rep. 2016;6:22190.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Rooney S, Alt FW, Lombard D, Whitlow S, Eckersdorff M, Fleming J, et al. Defective DNA repair and increased genomic instability in Artemis-deficient murine cells. J Exp Med. 2003;197(5):553–65.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Ziech D, Franco R, Pappa A, Panayiotidis MI. Reactive oxygen species (ROS)—induced genetic and epigenetic alterations in human carcinogenesis. Mutat Res. 2011;711(1–2):167–73.

    Article  PubMed  CAS  Google Scholar 

  122. Limoli CL, Giedzinski E, Morgan WF, Swarts SG, Jones GD, Hyun W. Persistent oxidative stress in chromosomally unstable cells. Cancer Res. 2003;63(12):3107–11.

    PubMed  CAS  Google Scholar 

  123. Morgan WF, Day JP, Kaplan MI, McGhee EM, Limoli CL. Genomic instability induced by ionizing radiation. Radiat Res. 1996;146(3):247–58.

    Article  PubMed  CAS  Google Scholar 

  124. Abdollahi H, Shiri I, Atashzar M, Sarebani M, Moloudi K, Samadian H. Radiation protection and secondary cancer prevention using biological radioprotectors in radiotherapy. Int J Cancer Ther Oncol. 2015;3(3):335.

    Article  Google Scholar 

  125. Kim K, Damoiseaux R, Norris AJ, Rivina L, Bradley K, Jung ME, et al. High throughput screening of small molecule libraries for modifiers of radiation responses. Int J Radiat Biol. 2011;87(8):839–45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  126. Xu G, Wu H, Zhang J, Li D, Wang Y, Wang Y, et al. Metformin ameliorates ionizing irradiation-induced long-term hematopoietic stem cell injury in mice. Free Radic Biol Med. 2015;87:15–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Zhang H, Zhai Z, Wang Y, Zhang J, Wu H, Wang Y, et al. Resveratrol ameliorates ionizing irradiation-induced long-term hematopoietic stem cell injury in mice. Free Radic Biol Med. 2013;54:40–50.

    Article  PubMed  CAS  Google Scholar 

  128. Li D, Tian Z, Tang W, Zhang J, Lu L, Sun Z, et al. The protective effects of 5-methoxytryptamine-α-lipoic acid on ionizing radiation-induced hematopoietic injury. Int J Mol Sci. 2016;17(6):935.

    Article  PubMed Central  CAS  Google Scholar 

  129. Van Buul JD, Fernandez-Borja M, Anthony EC, Hordijk PL. Expression and localization of NOX2 and NOX4 in primary human endothelial cells. Antioxid Redox Signal. 2005;7(3–4):308–17.

    Article  PubMed  Google Scholar 

  130. Han W, Wu L, Chen S, Bao L, Zhang L, Jiang E, et al. Constitutive nitric oxide acting as a possible intercellular signaling molecule in the initiation of radiation-induced DNA double strand breaks in non-irradiated bystander cells. Oncogene. 2007;26(16):2330–9.

    Article  PubMed  CAS  Google Scholar 

  131. Malaviya R, Gow AJ, Francis M, Abramova EV, Laskin JD, Laskin DL. Radiation-induced lung injury and inflammation in mice: role of inducible nitric oxide synthase and surfactant protein D. Toxicol Sci. 2014;144(1):27–38.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Nozaki Y, Hasegawa Y, Takeuchi A, Fan Z, Isobe K, Nakashima I, et al. Nitric oxide as an inflammatory mediator of radiation pneumonitis in rats. Am J Physiol Lung Cell Mol Physiol. 1997;272(4):L651–8.

    Article  CAS  Google Scholar 

  133. Tsuji C, Shioya S, Hirota Y, Fukuyama N, Kurita D, Tanigaki T, et al. Increased production of nitrotyrosine in lung tissue of rats with radiation-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol. 2000;278(4):L719–25.

    Article  PubMed  CAS  Google Scholar 

  134. Erbil Y, Dibekoglu C, Turkoglu U, Ademoglu E, Berber E, Kizir A, et al. Nitric oxide and radiation enteritis. Eur J Surg. 1998;164(11):863–8.

    Article  PubMed  CAS  Google Scholar 

  135. Abdollahi H, Atashzar M, Amini M. The potential use of biogas producing microorganisms in radiation protection. J Med Hypotheses Idea. 2015;9(2):67–71.

    Article  Google Scholar 

  136. Rwigema J-CM, Beck B, Wang W, Doemling A, Epperly MW, Shields D, et al. Two strategies for the development of mitochondrion-targeted small molecule radiation damage mitigators. Int J Radiat Oncol Biol Phys. 2011;80(3):860–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  137. Rajagopalan, Gupta K, Epperly MW, Franicola D, Zhang X, Wang H, et al. The mitochondria-targeted nitroxide JP4-039 augments potentially lethal irradiation damage repair. In Vivo (Athens, Greece). 2009;23(5):717–26.

    CAS  Google Scholar 

  138. Stoyanovsky DA, Huang Z, Jiang J, Belikova NA, Tyurin V, Epperly MW, et al. A manganese-porphyrin complex decomposes H(2)O(2), inhibits apoptosis, and acts as a radiation mitigator in vivo. ACS Med Chem Lett. 2011;2(11):814–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  139. Stoyanovsky D, Jiang J, Murphy M, Epperly M, Li S, Greenberger JS, et al. Mitigation of irradiation damage in vitro and in vivo by mitochondrial targeted glutathione peroxidase 4 mimic mito-ebselen. Int J Radiat Oncol Biol Phys. 2015;93(3):E541.

    Article  Google Scholar 

  140. Stoyanovsky DA, Jiang J, Murphy MP, Epperly M, Zhang X, Li S, et al. Design and synthesis of a mitochondria-targeted mimic of glutathione peroxidase, MitoEbselen-2, as a radiation mitigator. ACS Med Chem Lett. 2014;5(12):1304–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Aoyama T, Paik YH, Watanabe S, Laleu B, Gaggini F, Fioraso-Cartier L, et al. Nicotinamide adenine dinucleotide phosphate oxidase in experimental liver fibrosis: GKT137831 as a novel potential therapeutic agent. Hepatology. 2012;56(6):2316–27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Zykova TA, Zhu F, Zhai X, Ma WY, Ermakova SP, Lee KW, et al. Resveratrol directly targets COX-2 to inhibit carcinogenesis. Mol Carcinog. 2008;47(10):797–805.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Rwigema J-CM, Beck B, Wang W, Doemling A, Epperly MW, Shields D, et al. Two strategies for the development of mitochondrial-targeted small molecule radiation damage mitigators. Int J Radiat Oncol Biol Phys. 2011;80(3):860–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. Fetisova EK, Antoschina MM, Cherepanynets VD, Izumov DS, Kireev II, Kireev RI, et al. Radioprotective effects of mitochondria-targeted antioxidant SkQR1. Radiat Res. 2015;183(1):64–71.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to M. Najafi.

Ethics declarations

Conflict of interest

All authors declare that they have no conflict of interest.

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.

Disclaimer

All opinions are the personal and professional opinions of the authors and are not the opinions of their respective academic affiliations and agencies. Masoud Najafi and Vilmar Villa share both senior authorship.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yahyapour, R., Motevaseli, E., Rezaeyan, A. et al. Reduction–oxidation (redox) system in radiation-induced normal tissue injury: molecular mechanisms and implications in radiation therapeutics. Clin Transl Oncol 20, 975–988 (2018). https://doi.org/10.1007/s12094-017-1828-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12094-017-1828-6

Keywords

Navigation