Skip to main content
Wiley - PMC COVID-19 Collection logoLink to Wiley - PMC COVID-19 Collection
. 2001 Mar 16;11(2):87–101. doi: 10.1002/rmv.303

Role of free radicals in viral pathogenesis and mutation

Takaaki Akaike 1,
PMCID: PMC7169086  PMID: 11262528

Abstract

Oxygen radicals and nitric oxide (NO) are generated in excess in a diverse array of microbial infections. Emerging concepts in free radical biology are now shedding light on the pathogenesis of various diseases. Free‐radical induced pathogenicity in virus infections is of great importance, because evidence suggests that NO and oxygen radicals such as superoxide are key molecules in the pathogenesis of various infectious diseases. Although oxygen radicals and NO have an antimicrobial effect on bacteria and protozoa, they have opposing effects in virus infections such as influenza virus pneumonia and several other neurotropic virus infections. A high output of NO from inducible NO synthase, occurring in a variety of virus infections, produces highly reactive nitrogen oxide species, such as peroxynitrite, via interaction with oxygen radicals and reactive oxygen intermediates. The production of these various reactive species confers the diverse biological functions of NO. The reactive nitrogen species cause oxidative tissue injury and mutagenesis through oxidation and nitration of various biomolecules. The unique biological properties of free radicals are further illustrated by recent evidence showing accelerated viral mutation by NO‐induced oxidative stress. NO appears to affect a host's immune response, with immunopathological consequences. For example, NO is reported to suppress type 1 helper T cell‐dependent immune responses during infections, leading to type 2 helper T cell‐biased immunological host responses. NO‐induced immunosuppression may thus contribute to the pathogenesis of virus infections and help expansion of quasispecies population of viral pathogens. This review describes the pathophysiological roles of free radicals in the pathogenesis of viral disease and in viral mutation as related to both nonspecific inflammatory responses and immunological host reactions modulated by NO. Copyright © 2001 John Wiley & Sons, Ltd.

REFERENCES

  • 1. Granger DL, Hibbs JB Jr, Perfect JR, et al Specific amino acid (L‐arginine) requirement for microbiostatic activity of murine macrophages. J Clin Invest 1988; 81: 1129–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Nathan CF, Hibbs JB. Role of nitric oxide synthesis in macrophage antimicrobial activity. Curr Opin Immunol 1991; 3: 65–70. [DOI] [PubMed] [Google Scholar]
  • 3. Nathan C, Shiloh MU. Reactive oxygen and nitrogen intermediates in the relationship between mammalian hosts and microbial pathogens. J Clin Invest 2000; 97: 8841–8848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Doi T, Ando M, Akaike T, et al Resistance to nitric oxide in Mycobacterium avium complex and its implication in pathogenesis. Infect Immun 1993; 61: 1980–1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. James SL. Role of nitric oxide in parasitic infections. Microbiol Rev 1995; 59: 533–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Umezawa K, Akaike T, Fujii S, et al Induction of nitric oxide synthesis and xanthine oxidase and their role in the antimicrobial mechanism against Salmonella typhimurium in mice. Infect Immun 1997; 65: 2932–2940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Badwey JA, Karnovsky ML. Active oxygen species and the functions of phagocytic leukocytes. Annu Rev Biochem 1980; 49: 695–726. [DOI] [PubMed] [Google Scholar]
  • 8. Moncada S, Higgs A. The L‐arginine–nitric oxide pathway. N Engl J Med 1993; 329: 2002–2012. [DOI] [PubMed] [Google Scholar]
  • 9. Stuehr DJ, Griffith OW. Mammalian nitric oxide synthase. Adv Enzymol Relat Areas Mol Biol 1992; 65: 287–346. [DOI] [PubMed] [Google Scholar]
  • 10. Akaike T, Yoshida M, Miyamoto Y, et al Antagonistic action of imidazolineoxyl N‐oxides against endothelium‐derived relaxing factor/·NO through a radical reaction. Biochemistry 1993; 32: 827–832. [DOI] [PubMed] [Google Scholar]
  • 11. Maeda H, Akaike T. Oxygen free radicals as pathogenic molecules in viral diseases. Proc Soc Exp Biol Med 1991; 198: 721–727. [DOI] [PubMed] [Google Scholar]
  • 12. Akaike T, Suga M, Maeda H. Free radicals in viral pathogenesis: molecular mechanisms involving superoxide and NO. Proc Soc Exp Biol Med 1998; 217: 64–73. [DOI] [PubMed] [Google Scholar]
  • 13. Akaike T, Maeda H. Nitric oxide in influenza In Nitric Oxide in Infection, Fang FC. (ed.). Kluwer Academic/Plenum Publishers: New York, 1999; 397–415. [Google Scholar]
  • 14. Akaike T, Maeda H. Pathophysiological effects of high‐output production of nitric oxide In Nitric Oxide: Biology and Pathobiology, Ignarro LJ. (ed.). Academic Press: San Diego, CA, 2000; 733–745. [Google Scholar]
  • 15. Akaike T, Maeda H. Nitric oxide and virus infection. Immunology 2000; 101: 300–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Beckman JS, Beckman TW, Chen J, et al Apparent hydroxyl radical production by peroxynitrite: implications for endothelial injury from nitric oxide and superoxide. Proc Natl Acad Sci U S A 1990; 87: 1620–1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Beckman JS, Koppenol WH. Nitric oxide, superoxide and peroxynitrite: the good, the bad, and the ugly. Am J Physiol 1996; 271: C1424–1437. [DOI] [PubMed] [Google Scholar]
  • 18. Rubbo H, Darley‐Usmar V, Freeman BA. Nitric oxide regulation of tissue free radical injury. Chem Res Toxicol 1996; 9: 809–820. [DOI] [PubMed] [Google Scholar]
  • 19. Estévez AG, Crow JP, Sampson JB, et al Induction of nitric oxide‐dependent apoptosis in motor neurons by zinc‐deficient superoxide dismutase. Science 1999; 286: 2498–2500. [DOI] [PubMed] [Google Scholar]
  • 20. Sawa T, Akaike T, Maeda H. Tyrosine nitration by peroxynitrite formed from nitric oxide and superoxide generated by xanthine oxidase. J Biol Chem 2000; 275: 32467–32474. [DOI] [PubMed] [Google Scholar]
  • 21. Reiter CD, Teng RJ, Beckman JS. Superoxide reacts with nitric oxide to nitrate tyrosine at physiological pH via peroxynitrite. J Biol Chem 2000; 275: 32460–32466. [DOI] [PubMed] [Google Scholar]
  • 22. Szabó C, Ohshima H. DNA damage induced by peroxynitrite: subsequent biological effects. Nitric Oxide 1997; 1: 373–385. [DOI] [PubMed] [Google Scholar]
  • 23. Yermilov V, Rubio J, Ohshima H. Formation of 8‐nitroguanine in DNA treated with peroxynitrite in vitro and its rapid removal from DNA by depurination. Carcinogenesis 1995; 16: 2045–2050. [DOI] [PubMed] [Google Scholar]
  • 24. Rotrosen D, Gallin JI. Disorders of phagocyte function. Annu Rev Immunol 1987; 5: 127–150. [DOI] [PubMed] [Google Scholar]
  • 25. Nunoi H, Rotrosen D, Gallin JI, et al Two forms of autosomal chronic granulomatous disease lack distinct neutrophil cytosol factors. Science 1988; 242: 1298–1301. [DOI] [PubMed] [Google Scholar]
  • 26. Weiss J. Oxygen, ischemia and inflammation. Acta Physiol Scand Suppl 1986; 548: 9–37. [PubMed] [Google Scholar]
  • 27. Fridovich I. Superoxide radical and superoxide dismutases. Annu Rev Biochem 1995; 64: 97–112. [DOI] [PubMed] [Google Scholar]
  • 28. McCord JM. Oxygen‐derived free radicals in postischemic tissue injury. N Engl J Med 1985; 312: 159–163. [DOI] [PubMed] [Google Scholar]
  • 29. Akaike T, Ando M, Oda T, et al Dependence on O2 generation by xanthine oxidase of pathogenesis of influenza virus infection in mice radicals. J Clin Invest 1990; 85: 739–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Oda T, Akaike T, Hamamoto T, et al Oxygen radicals in influenza‐induced pathogenesis and treatment with pyran polymer‐conjugated SOD. Science 1989; 244: 974–976. [DOI] [PubMed] [Google Scholar]
  • 31. Ikeda T, Shimokata K, Daikoku T, et al Pathogenesis of cytomegalovirus‐associated pneumonitis in ICR mice: possible involvement of superoxide radicals. Arch Virol 1992; 127: 11–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Fridovich I. The biology of oxygen radicals. Science 1978; 201: 875–880. [DOI] [PubMed] [Google Scholar]
  • 33. Halliwell B, Gutteridge JMC. Oxygen toxicity, oxygen radicals, transition metals and disease. Biochem J 1984; 219: 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Peterhans E, Grob M, Bürge T, et al Virus‐induced formation of reactive oxygen intermediates in phagocytic cells. Free Radic Res Commun 1987; 3: 39–46. [DOI] [PubMed] [Google Scholar]
  • 35. Schwartz KB. Oxidative stress during viral infection: a review. Free Rad Biol Med 1996; 21: 641–649. [DOI] [PubMed] [Google Scholar]
  • 36. Bukrinsky MI, Nottet HSLM, Schmidtmayerova H, et al Regulation of nitric oxide synthase activity in human immunodeficiency virus type 1 (HSV‐1)‐infected monocytes: implications for HIV‐associated neurological disease. J Exp Med 1995; 181: 735–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Majano PL, García‐Monzón C, López‐Cabrera M, et al Inducible nitric oxide synthase expression in chronic viral hepatitis. Evidence for a virus‐induced gene upregulation. J Clin Invest 1998; 101: 1343–1352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Koprowski H, Zheng YM, Heber‐Katz E, et al In vivo expression of inducible nitric oxide synthase in experimentally induced neurologic diseases. Proc Natl Acad Sci U S A 1993; 90: 3024–3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Zheng YM, Schöfer MKH, Weihe E, et al Severity of neurological signs and degree of inflammatory lesions in the brains of the rats with Borna disease correlate with the induction of nitric oxide synthase. J Virol 1993; 67: 5786–5791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Karupiah G, Xie Q, Buller RML, et al Inhibition of viral replication by interferon‐γ‐induced nitric oxide synthase. Science 1993; 261: 1445–1448. [DOI] [PubMed] [Google Scholar]
  • 41. Akaike T, Weihe E, Schaefer M, et al Effect of neurotropic virus infection on neuronal and inducible nitric oxide synthase activity in rat brain. J Neurovirol 1995; 1: 118–125. [DOI] [PubMed] [Google Scholar]
  • 42. Mikami S, Kawashima S, Kanazawa K, et al Expression of nitric oxide synthase in a murine model of viral myocarditis induced by coxsackievirus B3. Biochem Biophys Res Commun 1996; 220: 983–989. [DOI] [PubMed] [Google Scholar]
  • 43. Akaike T, Noguchi Y, Ijiri S, et al Pathogenesis of influenza virus‐induced pneumonia: involvement of both nitric oxide and oxygen radicals. Proc Natl Acad Sci U S A 1996; 93: 2448–2453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Fujii S, Akaike T, Maeda H. Role of nitric oxide in pathogenesis of herpes simplex virus encephalitis in rats. Virology 1999; 256: 203–212. [DOI] [PubMed] [Google Scholar]
  • 45. Akaike T, Fujii S, Kato A, et al Viral mutation accelerated by nitric oxide production during infection in vivo . FASEB J 2000; 14: 1447–1454. [DOI] [PubMed] [Google Scholar]
  • 46. Cunha FQ, Moncada S, Liew FY. Interleukin‐10 (IL‐10) inhibits the induction of nitric oxide synthase by interferon‐γ in murine macrophages. Biochem Biophys Res Commun 1992; 182: 1155–1159. [DOI] [PubMed] [Google Scholar]
  • 47. Vodovotz Y, Bogdan C, Paik J, et al Mechanisms of suppression of macrophage nitric oxide release by transforming growth factor β. J Exp Med 1993; 178: 605–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Bogdan C, Vodovotz Y, Paik J, et al Mechanism of suppression of nitric oxide synthase expression by interleukin‐4 in primary mouse macrophages. J Leukoc Biol 1994; 55: 227–233. [DOI] [PubMed] [Google Scholar]
  • 49. Corraliza IM, Soler G, Eichmann K, et al Arginase induction by suppression of nitric oxide synthesis (IL‐4, IL‐10 and PGE2) in murine bone marrow‐derived macrophages. Biochem Biophys Res Commun 1995; 206: 667–673. [DOI] [PubMed] [Google Scholar]
  • 50. Gotoh T, Sonoki T, Nagasaki A, et al Molecular cloning of cDNA for nonhepatic mitochondrial arginase (arginase II) and comparison of its induction with nitric oxide synthase in a murine macrophage‐like cell line. FEBS Lett 1996; 395: 119–122. [DOI] [PubMed] [Google Scholar]
  • 51. Sonoki T, Nagasaki A, Gotoh T, et al Coinduction of nitric oxide synthase and arginase I in cultured rat peritoneal macrophages and rat tissues in vivo by lipopolysaccharide. J Biol Chem 1997; 272: 3689–3693. [DOI] [PubMed] [Google Scholar]
  • 52. Adamson DC, Kopnisky KL, Dawson TM, et al Mechanisms and structural determinants of HIV‐1 coat protein, gp41‐induced neurotoxicity. J Neurosci 1999; 19: 64–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Hori K, Burd PR, Furuke K, et al Human immunodeficiency virus‐1‐infected macrophages induce inducible nitric oxide synthase and nitric oxide (NO) production on astrocytes: astrocytic NO as a possible mediator of neuronal damage in acquired immunodeficiency syndrome. J Immunol 1999; 93: 1843–1850. [PubMed] [Google Scholar]
  • 54. Rostasy K, Monti L, Yiannoutsos C, et al Human immunodeficiency virus infection, inducible nitric oxide synthase expression, and microglial activation: pathogenetic relationship to the acquired immunodeficiency syndrome dementia complex. Ann Neurol 1999; 46: 207–216. [PubMed] [Google Scholar]
  • 55. Barbaro G, Di Lorenzo G, Soldini M, et al Intensity of myocardial expression of inducible nitric oxide synthase influences the clinical course of human immunodeficiency virus‐associated cardiomyopathy. Circulation 1999; 100: 933–939. [DOI] [PubMed] [Google Scholar]
  • 56. Tsutsumi H, Takeuchi R, Ohsaki M, et al Respiratory syncytial virus infection of human respiratory epithelial cells enhances inducible nitric oxide synthase gene expression. J Leukoc Biol 1999; 66: 99–104. [PubMed] [Google Scholar]
  • 57. Uetani K, Der SD, Zamanian‐Daryoush M, et al Central role of double‐stranded RNA‐activated protein kinase in microbial induction of nitric oxide synthase. J Immunol 2000; 165: 988–996. [DOI] [PubMed] [Google Scholar]
  • 58. Nathan CF. Inducible nitric oxide synthase: what difference does it make? J Clin Invest 1997; 100: 2417–2423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. MacMicking JD, North RJ, LaCourse R, et al Identification of nitric oxide synthase as a protective locus against tuberculosis. Proc Natl Acad Sci U S A 1997; 94: 5243–5248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Shiloh MU, MacMicking JD, Nicholson S, et al Phenotype of mice and macrophages deficient in both phagocyte oxidase and inducible nitric oxide synthase. Immunity 1999; 10: 29–38. [DOI] [PubMed] [Google Scholar]
  • 61. Shiloh MU, Nathan CF. Reactive nitrogen intermediates and the pathogensis of Salmonella and mycobacteria. Curr Opin Microbiol 2000; 3: 35–42. [DOI] [PubMed] [Google Scholar]
  • 62. Darrah PA, Hondalus MK, Chen Q, et al Cooperation between reactive oxygen and nitrogen intermediates in killing of Rhodococcus equi by activated macrophages. Infect Immun 2000; 68: 3587–3593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Mastroeni P, Vazquez‐Torres A, Fang FC, et al Antimicrobial actions of the NADPH phagocyte oxidase and inducible nitric oxide synthase in experimental salmonellosis. II. Effects on microbial proliferation and host survival in vivo . J Exp Med 2000; 192: 237–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Yoshida K, Akaike T, Doi T, et al Pronounced enhancement of ·NO‐dependent antimicrobial action by an ·NO‐oxidizing agent, imidazolineoxyl N‐oxide. Infect Immun 1993; 61: 3552–3555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. de Groote MA, Granger D, Xu Y, et al Genetic and redox determinants of nitric oxide cytotoxicity in a Salmonella typhimurium model. Proc Natl Acad Sci U S A 1995; 92: 6399–6403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Kuwahara H, Miyamoto Y, Akaike T, et al Helicobacter pylori urease suppresses bactericidal activity of peroxynitrite via carbon dioxide production. Infect Immun 2000; 68: 4378–4383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Miyamoto Y, Akaike T, Alam MS, et al Novel functions of human α1‐protease inhibitor after S‐nitrosylation: inhibition of cysteine protease and antibacterial activity. Biochem Biophys Res Commun 2000; 267: 918–923. [DOI] [PubMed] [Google Scholar]
  • 68. Stamler J, Singel D, Loscalzo J. Biochemistry of nitric oxide and its redox‐activated forms. Science 1992; 258: 1898–1902. [DOI] [PubMed] [Google Scholar]
  • 69. Akaike T. Mechanisms of biological S‐nitrosation and its measurement. Free Radic Res 2000; in press. [DOI] [PubMed] [Google Scholar]
  • 70. Croen KD. Evidence for an antiviral effect of nitric oxide. Inhibition of herpes simplex virus type 1 replication. J Clin Invest 1993; 91: 2446–2452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Mannick JB, Asano K, Izumi K, et al Nitric oxide produced by human B lymphocytes inhibits apoptosis and Epstein–Barr virus reactivation. Cell 1994; 79: 1137–1146. [DOI] [PubMed] [Google Scholar]
  • 72. Gao X, Tajima M, Sairenji T. Nitric oxide down‐regulates Epstein–Barr virus reactivation in epithelial cell lines. Virology 1999; 258: 375–381. [DOI] [PubMed] [Google Scholar]
  • 73. Saura M, Zaragoza C, McMillan A, et al An antiviral mechanism of nitric oxide: inhibition of a viral proteinase. Immunity 1999; 10: 21–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Karupiah G, Chen JH, Nathan CF, et al Identification of nitric oxide synthase 2 as an innate resistance locus against ectromelia virus infection. J Virol 1998; 72: 7703–7706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Zaragoza C, Ocampo CJ, Saura M, et al Inducible nitric oxide synthase protection against coxsackievirus pancreatitis. J Immunol 1999; 163: 5497–5504. [PubMed] [Google Scholar]
  • 76. van den Broek M, Bachmann MF, Höhler G, et al IL‐4 and IL‐10 antagonize IL‐12‐mediated protection against acute vaccinia virus infection with a limited role of IFN‐γ and nitric oxide synthetase 2. J Immunol 2000; 164: 371–378. [DOI] [PubMed] [Google Scholar]
  • 77. Karupiah G, Chen JH, Mahalingam S, et al Rapid interferon gamma‐dependent clearance of influenza A virus and protection from consolidating pneumonitis in nitric oxide synthase 2‐deficient mice. J Exp Med 1998; 188: 1541–1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Bartholdy C, Nansen A, Christensen JE, et al Inducible nitric‐oxide synthase plays a minimal role in lymphocytic choriomeningitis virus‐induced, T cell‐mediated protective immunity and immunopathology. J Gen Virol 1999; 80: 2997–3005. [DOI] [PubMed] [Google Scholar]
  • 79. Wu GF, Pewe L, Perlman S. Coronavirus‐induced demyelination occurs in the absence of inducible nitric oxide synthase. J Virol 2000; 74: 7683–7686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Guillemard E, Varano B, Belardelli F, et al Inhibitory activity of constitutive nitric oxide on the expression of alpha/beta interferon genes in murine peritoneal macrophages. J Virol 1999; 73: 7328–7333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Kreil TR, Eibl MM. Nitric oxide and viral infection: no antiviral activity against a flavivirus in vitro, and evidence for contribution to pathogenesis in experimental infection in vivo . Virology 1996; 219: 304–306. [DOI] [PubMed] [Google Scholar]
  • 82. Adler H, Beland JL, Del‐Pan NC, et al Suppression of herpes simplex virus type 1 (HSV‐1)‐induced pneumonia in mice by inhibition of inducible nitric oxide synthase (iNOS, NOS2). J Exp Med 1997; 185: 1533–1540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Nishio R, Matsumori A, Shioi T, et al Treatment of experimental viral myocarditis with interleukin‐10. Circulation 1999; 100: 1102–1108. [DOI] [PubMed] [Google Scholar]
  • 84. Hirasawa K, Jun HS, Hans HS, et al Prevention of encephalomyocarditis virus‐induced diabetes in mice by inhibition of the tyrosine kinase signaling pathway and subsequent suppression of nitric oxide production in macrophages. J Virol 1999; 73: 8541–8548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Andrews DM, Matthews VB, Sammels LM, et al The severity of Murray Valley encephalitis in mice is linked to neutrophil infiltration and inducible nitric oxide synthase activity in the central nervous system. J Virol 1999; 73: 8781–8790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Sidwell RW, Huffman JH, Bailey KW, et al Inhibitory effects of recombinant manganese superoxide dismutase on influenza virus infections in mice. Antimicrob Agents Chemother 1996; 40: 2626–2631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Lander HM. An essential role of free radicals and derived species in signal transduction. FASEB J 1997; 11: 118–124. [PubMed] [Google Scholar]
  • 88. Inoue K, Akaike T, Miyamoto Y, et al Nitrosothiol formation catalyzed by ceruloplasmin. Implication for cytoprotective mechanism in vivo . J Biol Chem 1999; 274: 27069–27075. [DOI] [PubMed] [Google Scholar]
  • 89. Ogura T, Tatemichi M, Esumi H. Nitric oxide inhibits CPP32‐like activity under redox regulation. Biochem Biophys Res Commun 1997; 236: 365–369. [DOI] [PubMed] [Google Scholar]
  • 90. Hortelano S, Alvarez AM, Bosca L. Nitric oxide induces tyrosine nitration and release of cytochrome c preceding an increase of mitochondrial transmembrane potential in macrophages. FASEB J 1999; 13: 2311–2317. [DOI] [PubMed] [Google Scholar]
  • 91. Okamoto T, Akaike T, Nagano T, et al Activation of human neutrophil procollagenase by nitrogen dioxide and peroxynitrite: a novel mechanism of procollagenase activation involving nitric oxide. Arch Biochem Biophys 1997; 342: 261–274. [DOI] [PubMed] [Google Scholar]
  • 92. Matsumoto H, Sies H. The reaction of ebselen with peroxynitrite. Chem Res Toxicol 1996; 9: 262–267. [DOI] [PubMed] [Google Scholar]
  • 93. Weitzman SA, Stossel TP. Mutation caused by human phagocytes. Science 1981; 212: 546–547. [DOI] [PubMed] [Google Scholar]
  • 94. Tsugita A, Fraenkel‐Conrat H. The composition of proteins of chemically evoked mutants of TMV RNA. J Mol Biol 1962; 4: 73–82. [DOI] [PubMed] [Google Scholar]
  • 95. Singer B, Fraenkel‐Conrat H. Mutagenicity of alkyl and nitroso‐alkyl compounds acting on tobacco mosaic virus and its RNA. Virology 1969; 39: 395–399. [DOI] [PubMed] [Google Scholar]
  • 96. Carp RI, Koprowski H. Mutation of type 3 poliovirus with nitrous acid. Virology 1962; 17: 99–109. [DOI] [PubMed] [Google Scholar]
  • 97. Granoff A. Induction of Newcastle disease virus mutants with nitrous acid. Virology 1961; 13: 402–408. [DOI] [PubMed] [Google Scholar]
  • 98. Holland JJ, Domingo E, de la Torre JC, et al Mutation frequencies at defined single codon sites in vesicular stomatitis virus and poliovirus can be increased only slightly by chemical mutagenesis. J Virol 1990; 64: 3960–3962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Juedes MJ, Wogan GN. Peroxynitrite‐induced mutation spectra of pSP189 following replication in bacteria and in human cells. Mutat Res 1996; 349: 51–61. [DOI] [PubMed] [Google Scholar]
  • 100. Zhuang JC, Lin C, Lin D, Wogan GN. Mutagenesis associated with nitric oxide production in macrophages. Proc Natl Acad Sci U S A 1998; 95: 8286–8291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Gal A, Wogan GN. Mutagenesis associated with nitric oxide production in transgenic SJL mice. Proc Natl Acad Sci U S A 1996; 93: 15102–15107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Calmels S, Hainaut P, Ohshima H. Nitric oxide induces conformational and functional modifications of wild‐type p53 tumor suppressor protein. Cancer Res 1997; 57: 3365–3369. [PubMed] [Google Scholar]
  • 103. Jaiswal M, LaRusso NF, Burgart LJ, et al Inflammatory cytokines induce DNA damage and inhibit DNA repair in cholangiocarcinoma cells by a nitric oxide‐dependent mechanism. Cancer Res 2000; 60: 184–190. [PubMed] [Google Scholar]
  • 104. Ames BN. Dietary carcinogens and anticarcinogens. Oxygen radicals and degenerative diseases. Science 1983; 221: 1256–1264. [DOI] [PubMed] [Google Scholar]
  • 105. Vuillaume M. Reduced oxygen species, mutation, induction and cancer initiation. Mutat Res 1987; 186: 43–72. [DOI] [PubMed] [Google Scholar]
  • 106. Harris CC. Chemical and physical carcinogenesis: advances and perspectives for the 1990s. Cancer Res 1991; 51: 5023s–5044s. [PubMed] [Google Scholar]
  • 107. Witz G. Active oxygen species as factors in multistage carcinogenesis. Proc Soc Exp Biol Med 1991; 198: 675–682. [DOI] [PubMed] [Google Scholar]
  • 108. Ames BN, Shigenaga MK, Hagen TM. Oxidants, antioxidants, and the degenerative diseases of aging. Proc Natl Acad Sci U S A 1993; 90: 7915–7922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Domingo E, Menendez‐Arias L, Holland JJ. RNA virus fitness. Rev Med Virol 1997; 7: 87–96. [DOI] [PubMed] [Google Scholar]
  • 110. Holland J, Spindler K, Horodyski F, et al Rapid evolution of RNA genomes. Science 1982; 215: 1577–1585. [DOI] [PubMed] [Google Scholar]
  • 111. Drake JW. Rates of spontaneous mutation among RNA viruses. Proc Natl Acad Sci U S A 1993; 90: 4171–4175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Drake JW, Charlesworth B, Charlesworth D, et al Rates of spontaneous mutation. Genetics 1998; 148: 1667–1686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Leider JM, Palese P, Smith FI. Determination of the mutation rate of a retrovirus. J Virol 1988; 62: 3084–3091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Petersen J, Dandri M, Burkle A, et al Increase in the frequency of hepadnavirus DNA integrations by oxidative DNA damage and inhibition of DNA repair. J Virol 1997; 71: 5455–5463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Beck MA, Shi Q, Morris VG, et al Rapid genomic evolution of a non‐virulent coxsackievirus B3 in selenium‐deficient mice results in selection of identical virulent isolates. Nat Med 1995; 1: 433–436. [DOI] [PubMed] [Google Scholar]
  • 116. Beck MA, Esworthy RS, Ho Y‐S, et al Glutathione peroxidase protects mice from viral‐induced myocarditis. FASEB J 1998; 12: 1143–1149. [DOI] [PubMed] [Google Scholar]
  • 117. Domingo E. Rapid evolution of viral RNA genomes. J Nutr 1997; 127: 958S–961S. [DOI] [PubMed] [Google Scholar]
  • 118. Smith DB, Inglis SC. The mutation rate and variability of eukaryotic viruses: an analytical review. J Gen Virol 1987; 68: 2729–2740. [DOI] [PubMed] [Google Scholar]
  • 119. Portner A, Webster RG, Bean WJ. Similar frequencies of antigenic variants in Sendai, vesicular stomatitis, and influenza A viruses. Virology 1980; 104: 235–238. [DOI] [PubMed] [Google Scholar]
  • 120. Nishikawa K, Isomura S, Suzuki S, et al Monoclonal antibodies of the HN glucoprotein of Newcastle disease virus. Biological characterization and use for strain comparisons. Virology 1983; 130: 318–330. [DOI] [PubMed] [Google Scholar]
  • 121. Kimata JT, Kuller L, Anderson DB, et al Emerging cytopathic and antigenic simian immunodeficiency virus variants influence AIDS progression. Nat Med 1999; 5: 535–541. [DOI] [PubMed] [Google Scholar]
  • 122. Taylor‐Robinson AW, Liew FY, Severn A, et al Regulation of the immune response by nitric oxide differentially produced by T helper type 1 and T helper type 2 cells. Eur J Immunol 1994; 24: 980–984. [DOI] [PubMed] [Google Scholar]
  • 123. Wei XQ, Charles IG, Smith A, et al Altered immune responses in mice lacking inducible nitric oxide synthase. Nature 1995; 375: 408–411. [DOI] [PubMed] [Google Scholar]
  • 124. Kolb H, Kolb‐Bachofen V. Nitric oxide in autoimmune disease: cytotoxic or regulatory mediator? Immunol Today 1998; 12: 556–561. [DOI] [PubMed] [Google Scholar]
  • 125. Zinkernagel RM. Immunology taught by viruses. Science 1996; 271: 173–178. [DOI] [PubMed] [Google Scholar]
  • 126. Bennink JR, Doherty PC. Different rules govern help for cytotoxic T cells and B cells. Nature 1978; 276: 829–831. [DOI] [PubMed] [Google Scholar]
  • 127. MacLean A, Wei XQ, Huang FP, et al Mice lacking inducible nitric‐oxide synthase are more susceptible to herpes simplex virus infection despite enhanced Th1 cell responses. J Gen Virol 1998; 79: 825–830. [DOI] [PubMed] [Google Scholar]
  • 128. Huang FP, Niedbala W, Wei XQ, et al Nitric oxide regulates Th1 cell development through the inhibition of IL‐12 synthesis by macrophages. Eur J Immunol 1998; 28: 4062–4070. [DOI] [PubMed] [Google Scholar]
  • 129. Mukhopadhyay S, George A, Bal V, et al Bruton's tyrosine kinase deficiency in macrophages inhibits nitric oxide generation leading to enhancement of IL‐12 induction. J Immunol 1999; 163: 1786–1792. [PubMed] [Google Scholar]
  • 130. Gherardi MM, Ramirez JC, Esteban M. Interleukin‐12 (IL‐12) enhancement of the cellular immune response against human immunodeficiency virus type 1 env antigen in a DNA prime/vaccinia virus boost vaccine regimen is time and dose dependent: suppressive effects of IL‐12 boost are mediated by nitric oxide. J Virol 2000; 74: 6278–6286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Ramsay AJ, Ruby J, Ramshaw IA. A case for cytokines as effector molecules in the resolution of virus infection. Immunol Today 1993; 14: 155–157. [DOI] [PubMed] [Google Scholar]
  • 132. Eichelberger M, Allan W, Zijlstra M, et al Clearance of influenza virus respiratory infection in mice lacking class I major histocompatibility complex‐restricted CD8+ T cells. J Exp Med 1991; 174: 875–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Graham MB, Braciale VL, Braciale TJ. Influenza virus‐specific CD4+ T helper type 2 T lymphocytes do not promote recovery from experimental virus infection. J Exp Med 1994; 180: 1273–1282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Graham MB, Dalton DK, Giltinan D, et al Response to influenza infection in mice with a targeted disruption in the interferon γ gene. J Exp Med 1993; 178: 1725–1732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Doherty TM, Sher A. Defects in cell‐mediated immunity affect chronic, but not innate, resistance of mice to Mycobacterium avium infection. J Immunol 1997; 158: 4822–4831. [PubMed] [Google Scholar]
  • 136. Vazquez‐Torres A, Jones‐Carson J, Mastroeni P, et al Antimicrobial actions of the NADPH phagocyte oxidase and inducible nitric oxide synthase in experimental salmonellosis. I. Effects on microbial killing by activated peritoneal macrophages in vitro . J Exp Med 2000; 192: 227–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. van der Vliet A, Eiserich JP, Shigenaga MK, et al Reactive nitrogen species and tyrosine nitration in the respiratory tract: epiphenomena or a pathobiologic mechanism of disease? Am J Respir Crit Care Med 1999; 160: 1–9. [DOI] [PubMed] [Google Scholar]
  • 138. Ignarro LJ. Introduction and overview In Nitric Oxide: Biology and Pathobiology, Ignarro, LJ (ed.). Academic Press: San Diego, CA, 2000; 3–19. [Google Scholar]

Articles from Reviews in Medical Virology are provided here courtesy of Wiley

RESOURCES