Skip to main content

Oxidatively Generated Damage to DNA and Biomarkers

  • Chapter
  • First Online:
Studies on Experimental Models

Abstract

The first part of this chapter is devoted to the description of degradation pathways for DNA bases and sugar moieties in the cell that are mediated by hydroxyl radical, one-electron oxidants and singlet oxygen. Thus, 11 single modified nucleosides and nucleobases that may be part of more complex radiation-induced DNA damage have been shown to be generated in nuclear DNA. In addition, four clustered DNA addition products and one tandem base-sugar lesion that arises from •OH-mediated hydrogen abstraction at C4 and C5 of the 2-deoxyribose moiety of DNA have also been identified in cells. Mechanisms of formation that are inferred from model studies are available for the 16 single and complex lesions thus detected in nuclear DNA. The DNA oxidation products whose radiation-induced formation in cellular was found to vary between 2 and 100 per 109 normal nucleosides per Gray may be used as biomarkers of oxidative stress. The accurate measurement of single, clustered or tandem lesions was performed, once DNA was extracted and subsequently enzymatically or chemically hydrolyzed, using accurate chromatographic methods. These involve, in most cases, the association of high performance liquid chromatography with the electrospray ionization tandem mass spectrometry (HPLC-ESI/MS–MS) detection techniques operating in the highly accurate and sensitive multiple reaction monitoring mode. Another approach that is more sensitive and less prone to artefactual oxidation consists of pre-incubating oxidized DNA with repair enzymes to reveal classes of modifications as strand breaks prior to either single-cell electrophoresis analysis or alkaline elution analysis.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 329.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Harman D (1981) The aging process. Proc Natl Acad Sci USA 78:7124–7128.

    Google Scholar 

  2. Floyd RA (1999) Neuroinflammatory processes are important in neurodegenerative diseases: an hypothesis to explain the increased formation of reactive oxygen and nitrogen species as major factors involved in neurodegenerative disease development. Free Radic Biol Med 26:1346–1355.

    Google Scholar 

  3. Bjelland S, Seeberg E (2003) Mutagenicity, toxicity and repair of DNA base damage induced by oxidation. Mutat Res 531:37–80.

    Google Scholar 

  4. Halliwell B (2006) Oxidative stress and neurodegeneration: where are we now? J Neurochem 97:1634–1658.

    Google Scholar 

  5. Halliwell B (2007) Oxidative stress and cancer: have we moved forward? Biochemical J 401:1–11.

    Google Scholar 

  6. Franco R, Schonevel O, Georgakilas AG, Panayiotidis MI (2008) Oxidative stress, DNA methylation and carcinogenesis. Cancer Lett 266:6–11.

    Google Scholar 

  7. Møller P, Løhr M, Folkmann JK, Mikkelsen L, Loft S (2010) Aging and oxidatively damaged nuclear DNA in animal organs. Free Radic Biol Med 48:1275–1285.

    Google Scholar 

  8. Sayre LM, Perry G, Mark A, Smith MA (2008) Oxidative stress and neurotoxicity. Chem Res. Toxicol 21:172–188.

    Google Scholar 

  9. Marnett LJ (2002) Oxy radicals, lipid peroxidation and DNA damage. Toxicology 181-182:219–222.

    Google Scholar 

  10. Misiaszek R, Crean C, Joffe A, Geacintov NE, Shafirovich V (2004) Oxidative DNA damage associated with combination of guanine and superoxide radicals and repair mechanisms via radical trapping. J Biol. Chem. 279:32106–32115.

    Google Scholar 

  11. Cadet J, Di Mascio P (2006) Peroxides in biological systems. In Functional Groups in Organic Chemistry – The Chemistry of Peroxides, Rappoport, Z. (ed.), Wiley and Sons, New York, Vol 2, pp. 915–1000.

    Google Scholar 

  12. Das AB, Nagy P, Abott HF, Winterbourn CC, Kettle AJ (2010) Reactions of superoxide with the myoglobin tyrosyl radical. Free Radic Biol Med 48:1540–1547.

    Google Scholar 

  13. Wagner JR, van Lier JE, Johnston LJ (1990) Quinone sensitized electron transfer photooxidation of nucleic acids: chemistry of thymine and thymidine radical cations in aqueous solution. Photochem Photobiol 52:333–343.

    Google Scholar 

  14. Frelon S, Douki T, Favier A, Cadet J (2002a) Comparative study of base damage induced by gamma radiation and Fenton reaction in isolated DNA. J Chem Soc Perkin Trans I 2866–2870.

    Google Scholar 

  15. Yamamoto K, Kawanishi S (1989) Hydroxyl free radical is not the main active species in site-specific DNA damage induced by copper (II) ion and hydrogen peroxide. J Biol Chem 264:15435–15440.

    Google Scholar 

  16. Frelon S, Douki T, Favier A, Cadet J (2003) Hydroxyl radical is not the main reactive species involved in the degradation of DNA bases by copper in the presence of hydrogen peroxide. Chem Res Toxicol 16:191–197.

    Google Scholar 

  17. Park J-H, Troxel, AB, Harvey RG, Penning TM 2006) Polycyclic aromatic hydrocarbon (PAH) o-quinones produced by the aldo-quinones produced by the aldo-keto-reductases (AKRs) generate abasic sites, oxidized pyrimidines and 8-oxo-dGuo via reactive oxygen species. Chem Res Toxicol 19:719–728.

    Google Scholar 

  18. Joffe A, Geacintov NE, Shafirovich V (2003) DNA lesions derived from the site selective oxidation of guanine by carbonate radical anions. Chem Res Toxicol 16:1528–1538.

    Google Scholar 

  19. Medinas DB, Cerchiaro G, Trindale DF, Augusto O (2007) The carbonate radical and related oxidants derived from bicarbonate buffer. IUBMB Life 59:255–262.

    Google Scholar 

  20. Mothersill C, Seymour CB (2004) Radiation-induced bystander effects – implication for cancer. Nat Rev Cancer 4:158–164.

    Google Scholar 

  21. Prise KM, O’Sullivan JM (2009) Radiation-induced bystander signaling in cancer therapy. Nat Rev Cancer 9:351–360.

    Google Scholar 

  22. Bagdonas S, Dahlie J, Kaalhus O, Moan J (1999) Cooperative inactivation of cells in microcolonies treated with UVA radiation. Radiat Res 152:174–179.

    Google Scholar 

  23. Alexandre J, Hu Y, Lu W, Pelicano H, Huang P (2007) Novel action of paditaxel against cancer cells: bystander effect mediated by reactive oxygen species. Cancer Res 67:3512–3517.

    Google Scholar 

  24. Grand A, Morell C, Labet V, Cadet J, Eriksson LA (2007) •H atom and •OH reactions with 5-methylcytosine. J Phys Chem A 111:8968–8972.

    Google Scholar 

  25. Munk BH, Burrows CJ, Schlegel HB (2007) Exploration of mechanisms for the transformation of 8-hydroxyguanine radical to FAPyG by density functional theory. Chem Res Toxicol 20:432–444.

    Google Scholar 

  26. Munk BH, Burrows CJ, Schlegel HB (2008) An exploration of mechanisms for the transformation of 8-oxoguanine to guanidinohydantoin and spiroiminodihydantoin by density functional theory. J Am Chem Soc 130:5245–5256.

    Google Scholar 

  27. Labet V, Grand A, Cadet J, Eriksson LA (2008) Deamination of the radical cation of base moiety of 2′-deoxycytidine: a theoretical study. Chemphyschem 9:1195–1203.

    Google Scholar 

  28. Labet V, Morell C, Grand A, Cadet J, Cimino P, Barone V (2008) Formation of cross-linked adducts between guanine and thymine mediated by hydroxyl radical and one-electron oxidation: a theoretical study. Org Biomol Chem 6:3300–3305.

    Google Scholar 

  29. Cadet J, Douki T, Gasparutto D, Ravanat J-L (2003) Oxidative damage to DNA: Formation, measurement and biochemical features. Mutat Res 531:5–23.

    Google Scholar 

  30. Cadet J, Ravanat J-L, Martinez GR, Medeiros MHG, Di Mascio P (2006) Singlet oxygen oxidation of isolated and cellular DNA: product formation and mechanistic insights. Photochem Photobiol 82:1219–1225.

    Google Scholar 

  31. Cadet J, Douki T, Ravanat J-L (2008) Oxidatively generated damage to the guanine moiety of DNA: Mechanistic aspects and formation in cells. Acc Chem Res 41:1075–1083.

    Google Scholar 

  32. Douki T, Ravanat J-L, Angelov D, Wagner JR, Cadet J (2004) Effects of duplex stability on charge transfer efficiency within DNA. Topics Curr Chem 236:1–25.

    Google Scholar 

  33. Gimisis T, Cismas C (2006) Isolation, characterization, and independent synthesis of guanine oxidation products. Eur J Org Chem 1351–1378.

    Google Scholar 

  34. Neeley WL, Essigmann JM (2006) Mechanisms of formation, genotoxicity, and mutation of guanine oxidation products. Chem Res Toxicol 19:491–505.

    Google Scholar 

  35. Pratviel G, Meunier B (2006) Guanine oxidation: one- and two-electron reactions. Chem Eur J 12:6018–6030.

    Google Scholar 

  36. Wagner JR, Cadet J (2010) Oxidation reactions of cytosine DNA components by hydroxyl radical and one-electron oxidants in aerated aqueous solutions. Acc Chem Res 43:563–571.

    Google Scholar 

  37. von Sonntag C, ed (2006) Free-radical induced DNA damage and its repair. A chemical perspective, Springer: Heidelberg, 2006.

    Google Scholar 

  38. Dedon PC (2008) The chemical toxicology of 2-deoxyribose oxidation in DNA. Chem Res Toxicol 21:206–219.

    Google Scholar 

  39. Chen B, Zhou X, Taghizadeh K, Chen, J, Stubbe J, Dedon PC (2007) CG-MS methods to quantify the 2-deoxypentos-4-ulose and 3′-phosphoglycolate pathways of 4′-oxidation of deoxyribose: Application to DNA damage produced by γ–irradiation and bleomycin. Chem Res Toxicol 20:1701–1708.

    Google Scholar 

  40. Douki T, Ravanat J-L, Pouget J-P, Testard I, Cadet J (2006) Minor contribution of direct ionization to DNA bas damage induced by heavy ions. Int J Radiat Biol 82:119–127.

    Google Scholar 

  41. Cadet J, Vigny P (1990) The photochemistry of nucleic acids. In: Bioorganic Photochemistry, Morrison, H. (ed.) Vol. 1, Wiley and Sons, New York, pp. 1–272.

    Google Scholar 

  42. Ballmaier D, Epe B (2006) DNA damage by bromate: mechanism and consequences. Toxicology 221, 166–171.

    Google Scholar 

  43. Douki T, Angelov D, Cadet J (2001) UV laser photolysis of DNA: effect of duplex stability on charge transfer efficiency. J Am Chem Soc 123:11360–11366.

    Google Scholar 

  44. Pouget J-P, Ravanat J-L, Douki T, Richard M-J, Cadet J (1999) Measurement of DNA base damage in cells exposed to low doses of gamma radiation: comparison between the HPLC/ECD and the comet assays. Int J Radiat Biol 75:51–58.

    Google Scholar 

  45. Pouget J-P, Frelon,S, Ravanat J-L, Testard I, Odin F, Cadet J (2002) Formation of modified DNA to DNA in cells exposed to either gamma radiation or high-LET particles. Radiat Res 157:589–595.

    Google Scholar 

  46. Frelon S, Douki T, Ravanat J-L, Pouget J-P, Tornabene C, Cadet J (2000) High-performance liquid chromatography-tandem mass spectrometry measurement of radiation-induced base damage to isolated and cellular DNA. Chem Res Toxicol 13:1002–1010.

    Google Scholar 

  47. Cadet J, Téoule R (1975) Radiolyse gamma de la thymidine en solution aqueuse aérée. 1. - Identification des hydroperoxydes. Bull Soc Chim Fr 879–884.

    Google Scholar 

  48. Wagner JR, van Lier JE, Berger M, Cadet J (1994) Thymidine hydroperoxides. Structural assignment, conformational features, and thermal decomposition in water. J Am Chem Soc 116:2235–2242.

    Google Scholar 

  49. Lutsig MJ, Cadet J, Boorstein RJ, Teebor GW (1992) Synthesis of the diastereomers of thymidine glycol, determination of concentrations and rates of interconversion of their cis-trans epimers at equilibrium and demonstration of differential alkali lability within DNA. Nucleic Acids Res 20:4839–4845.

    Google Scholar 

  50. Douki T, Delatour T, Paganon F, Cadet J (1996) Measurement of oxidative damage at pyrimidine bases in gamma-irradiated DNA. Chem Res Toxicol 9:1145–1151.

    Google Scholar 

  51. Decarroz C, Wagner JR, van Lier JE, Krishna CM, Riesz P, Cadet J (1986) Sensitized photo-oxidation of thymidine by 2-methyl-1,4-naphthoquinone. Characterization of the stable photoproducts. Int J Radiat Biol 50:491–505.

    Google Scholar 

  52. Steenken S (1989) Purine bases, nucleosides and nucleotides: Aqueous solution redox chemistry and transformation reactions of radical cations, e- and OH adducts. Chem Rev 89:503–520.

    Google Scholar 

  53. Cadet J, Douki T, Ravanat J-L (2010) Oxidatively generated base damage to cellular DNA Free Radic Biol Med 49:9–21.

    Google Scholar 

  54. Bourdat AG, Douki T, Frelon S, Gasparutto D, Cadet J (2000) Tandem base lesions are generated by hydroxyl radical within isolated DNA in aerated aqueous solution. J Am Chem Soc 122:4549–4556.

    Google Scholar 

  55. Douki T, Rivière J, Cadet J (2002) DNA tandem lesions containing 8-oxo-7,8-dihydroguanine and formamide residues arise from intramolecular addition of thymine peroxyl radical to guanine. Chem Res Toxicol 15:445–454.

    Google Scholar 

  56. Cadet J, Berger M, Buchko GW, Joshi P, Raoul S, Ravanat J-L (1994) 2,2-Diamino-4-[(3,5-di-O-acetyl-2′-deoxy-β-D-erythro-pentofuranosyl)amino]-5-(2H)-oxazolone. J Am Chem Soc 116:7403–7404.

    Google Scholar 

  57. Raoul S, Berger M, Buchko GW, Joshi PC, Morin B, Weinfeld M, Cadet J (1996) 1H, 13C and 15N NMR analysis and chemical features of the two main radical oxidation products of 2′-deoxyguanosine: oxazolone and imidazolone nucleosides. J Chem Soc Perkin Trans 2 371–381.

    Google Scholar 

  58. Douki T, Cadet J (1999) Modification of DNA bases by photosensitized one-electron oxidation. Int J Radiat Biol 75:571–581.

    Google Scholar 

  59. Matter B, Malejka-Giganti D, Csallany AS, Tretyakova N (2006) Quantitative analysis of the oxidative DNA lesion, 2,2-diamino-4-(2-deoxy-beta-D-erythro-pentofuranosyl)amino]-5(2H)-oxazolone (oxazolone), in vitro and in vivo by isotope dilution-capillary HPLC-ESI-MS/MS. Nucleic Acids Res 34:5449–5460.

    Google Scholar 

  60. Chatgilialoglu C, D’Angelantonio M, Guerra M, Kaloudis P, Mulazzani QG (2009) A reevaluation of the ambient reactivity of the guanine moiety towards hydroxyl radicals. Angew Chem Int Ed 48:2214–2217.

    Google Scholar 

  61. Vialas C, Pratviel G, Claporols C, Meunier B (1998) Efficient oxidation of 2′-deoxyguanosine by Mn-TMPyP/KHSO5 to imidazolone dIz without formation of 8-Oxo-dG. J Am Chem Soc 120:11548–11553.

    Google Scholar 

  62. Kasai H, Yamaizumi Z, Berger M, Cadet J (1992) Photosensitized formation of 7,8-dihydro-8-oxo-2′-deoxyguanosine (8-hydroxy-2′-deoxyguanosine) in DNA by riboflavin: a non singlet oxygen mediated reaction. J Am Chem Soc 114:9692–9694.

    Google Scholar 

  63. Genereux JC, Barton JK (2010) Mechanisms for DNA charge transport. Chem Rev 110:1642–1662.

    Google Scholar 

  64. Kanvah S, Joseph J, Schuster GB, Barnett RN, Cleveland CL, Landman U (2010) Oxidation of DNA: damage to nucleobases. Acc Chem Res 43:280–287.

    Google Scholar 

  65. Berger M, de Hazen M, Nejjari, A, Fournier J, Guignard J, Pezerat H, Cadet J (1993) Radical oxidation reactions of the purine moiety of 2′-deoxyribonucleosides and DNA by iron-containing minerals. Carcinogenesis 14:41–46.

    Google Scholar 

  66. Raoul S, Bardet M, Cadet, J (1995) Gamma irradiation of 2′-deoxyadenosine in oxygen-free aqueous solutions: Identification and conformational features of formamidopyrimidine nucleoside derivatives. Chem Res Toxicol 8:924–933.

    Google Scholar 

  67. Bergeron F, Auvré F, Radicella JP, Ravanat J-L (2010) HO• radicals induce an unexpected high proportion of tandem base lesions refractory to repair by DNA glycosylases. Proc Natl Acad Sci USA 107:5528–5533.

    Google Scholar 

  68. Mori T, Dizdaroglu M (1994) Ionizing radiation causes greater DNA base damage in ­radiation-sensitive mutant M10 cells than in parent mouse lymphoma L5178Y cells. Radiat Res 140:65–90.

    Google Scholar 

  69. Mori T, Hori Y, Dizdaroglu M (1993) DNA base damage generated in vivo in hepatic ­chromatin of mice upon whole body γ–irradiation. Int J Radiat Biol 64:645–650.

    Google Scholar 

  70. Frelon S, Douki T, Cadet J (2002) Radical oxidation of the adenine moiety of nucleoside and DNA: 2-hydroxy-2′-deoxyadenosine is a minor decomposition product. Free Radic. Res. 36:499–508.

    Google Scholar 

  71. Gradzka I (2005) A non-radioactive, PGFE-based assay for low levels of DNA double-strand breaks in mammalian cells. DNA Repair (Amst.) 4:1129–1139.

    Google Scholar 

  72. Olive PL (2009) Impact of the comet assay in radiobiology. Mutat Res 681, 13–23.

    Google Scholar 

  73. Muslimovic A, Ismail IH, Gao Y, Hammarsten O (2008) An optimized method for measurement of gamma-H2AX in blood mononuclear and cultured cells. Nat Protocol 3, 1187–1193.

    Google Scholar 

  74. Box HC, Budzinski EE, Freund HG, Evans MS, Patrzyc HB, Wallace JC, MacCubbin AE (1993) Vicinal lesions in X-irradiated DNA. Int J Radiat Biol 64:261–263.

    Google Scholar 

  75. Box HC, Freund HG, Budzinski EE, Wallace JC, MacCubbin AE (1995) Free radical-induced double base lesions. Radiat Res 141, 91–94.

    Google Scholar 

  76. Hong IS, Carter KN, Sato K, Greenberg MM (2007a) Characterization and mechanism of formation of tandem lesions in DNA by a nucleobase peroxyl radical. J Am Chem Soc 129:4089–4098.

    Google Scholar 

  77. Hong H, Cao H, Wang Y (2007) Formation and genotoxicity of a guanine-cytosine intrastrand cross-link lesion in vivo. Nucleic Acids Res 35:7118–7127.

    Google Scholar 

  78. Ghosh A, Joy A, Schuster GB, Douki T, Cadet J (2008) Selective one-electron oxidation of duplex DNA oligomers: reactions at thymines. Org Biomol Chem 6:916–928.

    Google Scholar 

  79. Jiang Y, Hong H, Cao H, Wang Y (2007) In vivo formation and in vitro replication of a guanine-thymine intrastrand cross-link lesion. Biochemistry 46:12757–12763.

    Google Scholar 

  80. Cao H, Wang Y (2007) Quantification of oxidative single-base and intrastrand cross-link lesions in unmethylated and CpG-methylated DNA induced by Fenton-type reagents. Nucleic Acids Res 35:4833–4844.

    Google Scholar 

  81. Wang Y (2008) Bulky DNA lesions induced by reactive oxygen species. Chem Res Toxicol 21:276–281.

    Google Scholar 

  82. Zhang Q, Wang Y (2003) Independent generation of 5-(2′-deoxycytidinyl)methyl radical and the formation of a novel crosslink lesion between 5-methylcytosine and guanine. J Am Chem Soc 125:12795–12802.

    Google Scholar 

  83. Zhang Q, Wang Y (2005) Generation of 5-(2′-deoxycytidyl)methyl radical and the formation of intrastrand cross-link lesions in oligodeoxyribonucleotides. Nucleic Acids Res 33:1593–1603.

    Google Scholar 

  84. Romieu A, Bellon S, Gasparutto D, Cadet J (2000) Synthesis and UV photolysis of oligodeoxynucleotides that contain 5-(phenyl-thiomethyl)-2′-deoxyuridine: a specific photolabile precursor of 5-(2′-deoxyuridilyl)methyl radical. Org Lett 2:1085–1088.

    Google Scholar 

  85. Bellon S, Ravanat J-L, Gasparutto D, Cadet J (2002) Cross-linked thymine-purine base tandem lesions: synthesis, characterization, and measurement in gamma-irradiated DNA. Chem Res Toxicol 15:598–606.

    Google Scholar 

  86. Bellon S, Gasparutto D, Saint-Pierre C, Cadet J (2006) Guanine-thymine intrastrand cross-linked lesion containing oligonucleotides: from chemical synthesis to in vitro enzymatic replication. Org Biomol Chem 4:3831–3837.

    Google Scholar 

  87. Hong H, Cao H, Wang Y (2006) Identification and quantification of a guanine-thymine instrastrand cross-link lesion induced by Cu(II)H2O2/ascorbate. Chem Res Toxicol 19:614–621.

    Google Scholar 

  88. Xerri B, Morell C, Grand A, Cadet J, Cimino P, Barone V (2006) Radiation-induced formation of DNA intrastrand crosslinks between thymine and adenine bases. A theoretical approach. Org Biomol Chem 4:3986–3992.

    Google Scholar 

  89. Gu C, Wang Y (2004) LC-MS./MS identification and yeast polymerase eta bypass of a novel gamma-irradiation-induced intrastrand cross-link lesion G[8-5]C. Biochemistry 43:6745–6750.

    Google Scholar 

  90. Chatgilialoglu C, Gimisis T (1998) Fate of the C-1′ peroxyl radical in the 2′-deoxyuridine system. Chem Commun 1249–1259.

    Google Scholar 

  91. Regulus P, Spessotto S, Gateau M, Cadet J, Favier A, Ravanat J-L (2004) Detection of new radiation-induced DNA degradation lesions by liquid chromatography coupled to tandem mass spectrometry. Rapid Commun Mass Spectrom 18:2223–2228.

    Google Scholar 

  92. Regulus P, Duroux B, Bayle P-A, Favier A, Cadet J, Ravanat J-L (2007) Oxidation of the sugar moiety of DNA by ionizing radiation or bleomycin could induce the formation of a cluster DNA lesion. Proc Natl Acad Sci USA 104:14032–14037.

    Google Scholar 

  93. Dedon PC, Goldberg IH (1992) Free-radical mechanism involved in the formation of sequence-dependent bistranded DNA lesions by the antitumor antibiotics bleomycin, neocarzinostatin, and calicheamicin. Chem Res Toxicol 5:311–322.

    Google Scholar 

  94. Pogozelski WK, Tullius TD (1998) Oxidative strand scission of nucleic acids: routes initiated by hydrogen abstraction from the sugar moiety. Chem Rev 98:1089–1107.

    Google Scholar 

  95. Sczepanski JT, Jacobs AC, Greenberg MM (2008) Self-promoted interstrand cross-link ­formation by an abasic site. J Am Chem Soc 130:9646–9647.

    Google Scholar 

  96. Sczepanski JT, Jacobs AC, Majumdar A, Greenberg MM (2009) Scope and mechanism cross-link formation by the C4’-oxidized abasic site. J Am Chem Soc 131:11132–11139.

    Google Scholar 

  97. Keck K (1968) Bildung von cyclonucleotiden bei bestrahlung wassiriger losungen von purinnucleotiden. Z Naturforsch 23b:1034–1043.

    Google Scholar 

  98. Mariaggi N, Cadet J, Téoule R (1976) Cyclisation radicalaire de la désoxy-2′-adénosine en solution aqueuse, sous l’effet du rayonnement gamma. Tetrahedron 32:2385–2387.

    Google Scholar 

  99. Raleigh JA, Fuciarelli AF (1985) Distribution of damage in irradiated 5′-AMP: 8,5′-cyclo-AMP, 8-hydroxy-AMP, and adenine release. Radiat Res 102:65–175.

    Google Scholar 

  100. Dizdaroglu M (1986a) Free-radical-induced formation of an 8,5′-cyclo-2′-deoxyguanosine moiety in deoxyribonucleic acid. Biochem J 238:247–254.

    Google Scholar 

  101. Belmadoui N, Boussicault F, Guerra M, Ravanat J-L, Chatgilialoglu C, Cadet J 2010 Org Biomol Chem 8:3211–3219.

    Google Scholar 

  102. Jaruga P, Xian Y, Nelson BC, Dizdaroglu M (2009) Measurement of (5′R)- and (5′S)-8,5′-cyclo-2′-deoxyadenosines in DNA in vivo by liquid chromatography/isotope-dilution tandem mass spectrometry. Biochem Biophys Res Commun 386:656–666.

    Google Scholar 

  103. Manetto A, Georganakis D, Leondiadis L, Gimisis T, Mayer P, Carell T, Chatgilialoglu C (2007) Independent generation of C5′-nucleosidyl radicals in thymidine and 2′-deoxyguanosine. J Org Chem 72:3659–3666.

    Google Scholar 

  104. Chatgilialoglu C, Guerra M, Mulazzani QG (2003) Model studies of DNA C5′ radicals. Selective generation and reactivity of 2′-deoxyadenosin-5′-yl radical. J Am Chem Soc 125:3839–3848.

    Google Scholar 

  105. D’Errico M, Parlanti E, Teson M, Bernades de Jesus BM, Degan P, Calcagnile A, Jaruga P, Bjørås M, Crescenzi M, Pedrini AM, Egly J-M, Zambruno G, Stefanini M, Dizdaroglu M, Dogliotti E (2006) New functions of XPC in the protection of human skin cells from oxidative damage. EMBO J 25:4305–4315.

    Google Scholar 

  106. Cadet J, Poulsen H (2010) Measurement of oxidatively generated base damage in cellular DNA and urine. Free Radic Biol Med 48:1457–1459.

    Google Scholar 

  107. Sheu C, Foote CS (1993) Endoperoxide formation in a guanosine derivative. J Am Chem Soc 115:10446–10447.

    Google Scholar 

  108. Sheu C, Kang P, Khan S, Foote CS (2002) Low-temperature photosensitized oxidation of a guanosine derivative and formation of an imidazole ring-opened product. J Am Chem Soc 124:3905–3913.

    Google Scholar 

  109. Niles JC, Wishnok JS, Tannenbaum SR (2001) Spiroiminodihydantoin is the major product of 8-oxo-7,8-dihydroguanosine with peroxynitrite in the presence of thiols and guanosine oxidation by methylene blue. Org Lett 3:963–936.

    Google Scholar 

  110. Martinez GR, Ravanat J-L, Medeiros MHG, Cadet J, Di Mascio P (2000) Synthesis of a naphthalene endoperoxide as a source of 18O-labeled singlet oxygen for mechanistic studies. J Am Chem Soc 122:10212–10213.

    Google Scholar 

  111. Ravanat JL, Di Mascio P, Martinez GR, Medeiros MH, Cadet J (2000) Singlet oxygen induces oxidation of cellular DNA. J Biol Chem 275:40601–40604.

    Google Scholar 

  112. Cadet J, Douki T, Sage E (2005) Ultraviolet radiation-mediated damage to cellular DNA. Mutat Res 571:3–17.

    Google Scholar 

  113. Pouget J-P, Douki T, Richard M-J, Cadet J (2000) DNA damage induced in cells by γ and UVA radiation as measured by HPLC/GC-MS and HPLC-EC and comet assay. Chem Res Toxicol 13:541–549.

    Google Scholar 

  114. Mouret S, Baudouin C, Charveron M, Favier A, Cadet J, Douki T (2006) Cyclobutane pyrimidine dimers are predominant DNA lesions in whole human skin exposed to UVA radiation. Proc Natl Acad Sci USA 103:13765–13770.

    Google Scholar 

  115. Ravanat J-L, Sauvaigo S, Caillat S, Martinez GR, Medeiros MHG, Di Mascio P, Favier A, Cadet J (2004) Singlet-oxygen-mediated damage to cellular DNA determined by the comet assay associated with DNA repair enzymes. Biol Chem 385:7–20.

    Google Scholar 

  116. Hariharan PV, Cerutti PA (1972) Formation and repair of gamma-ray induced thymine damage in Micrococcus radiodurans. J Mol Biol 66:65–81.

    Google Scholar 

  117. Cadet J, Berger M (1985) Radiation-induced degradation of the purine bases within DNA and realted model compounds. Int J Radiat Biol 47:127–143.

    Google Scholar 

  118. Möller L, Hofer T (1997) [32P]ATP mediates formation of 8-hydroxy-2′-deoxyguanosine from 2′-deoxyguanosine, a possible problem in the 32P-postlabeling assay. Carcinogenesis 18:2415–2419.

    Google Scholar 

  119. Cadet J

    Google Scholar 

  120. Maynard S, de Souza-Pinto NC, Scheibye-Knudsen M, Bohr VA (2010) Mitochondrial base excision repair assays. Methods 51:416–425.

    Google Scholar 

  121. Yin B, Whyatt RM, Perer FP, Randall MC, Cooper TB, Santella RM (1995) Determination of 8-hydroxydeoxyguanosine by an immunoaffinity chromatography-monoclonal antibody-based ELISA. Free Radic Biol Med 18:1023–1032.

    Google Scholar 

  122. Toyokuni S, Tanaka T, Hattori Y, Nishiyama Y, Yoshida A, Uchida K, Hiai H, Ochi H, Osawa T (1997) Quantitative immunohistochemical determination of 8-hydroxy-2′-deoxyguanosine by a monoclonal antibody N45.1: its application to ferric nitrilotriacetate-induced renal carcinogenesis model. Lab Invest 76:365–374.

    Google Scholar 

  123. Song M-F, Li Y-S, Ootsuyama Y, Kasai H, Kawai K, Ohta M, Eguchi Y, Yamato M, Matsumoto Y, Yoshida, R.; Ogawa Y (2009) Urea, the most abundant component in urine, cross-reacts with a commercial 8-OHdG ELISA kit and contributes to over-estimation of urinary 8-OHdG. Free Radic Biol Med 47:41–46.

    Google Scholar 

  124. Garratt LW, Mistry V, Singh R, Sandhu J, Sheil B, Cooke MS, Sly PD (2010) Interpretation of urinary 8-oxo-7,8-dihydro-2′-deoxyguanosine is adversely affected by methodological inaccuracies when using a commercial ELISA. Free Radic Biol Med 48:1464–1468.

    Google Scholar 

  125. Dizdaroglu M (1986) Characterization of free-radical induced damage to DNA by the combined use of enzymatic hydrolysis and gas-chromatography-mass spectrometry. J Chromatogr 367:357–366.

    Google Scholar 

  126. Dizdaroglu M, Dirksen ML, Jiang HX, Robbins JH (1987) Ionizing-radiation-induced damage in the DNA of cultured human cells: identification of 8,5-cyclo-2-deoxyguanosine. Biochem J 241:929–932.

    Google Scholar 

  127. Dizdaroglu M, Nackerdien Z, Chao BC, Gajewski E, Rao G (1991) Chemical nature of in vivo DNA damage in hydrogen peroxide-treated mammalian cells. Arch Biochem Biophys 285:388–390.

    Google Scholar 

  128. Floyd RA, Watson JJ, Wong PK, Altmiller DH, Rickard RC (1986) Hydroxyl free radical adduct of deoxyguanosine: sensitive detection and mechanism of formation. Free Radic Res Commun 1:163–172.

    Google Scholar 

  129. Kasai H, Nishimura S (1984) Hydroxylation of deoxyguanosine at the C-8 position by ascorbic acid and other reducing agents. Nucleic Acids Res (1984) 12:2137–2145.

    Google Scholar 

  130. Kasai H (1997) Analysis of a form of oxidative DNA damage, 8-hydroxy-2′-deoxyguanosine, as a marker of cellular oxidative stress during carcinogenesis. Mutat Res 387:147–163.

    Google Scholar 

  131. Halliwell B, Dizdaroglu M (1992) The measurement of oxidative damage to DNA by HPLC and GC-MS techniques. Free Radic Res Commun 16:75–87.

    Google Scholar 

  132. Ravanat J-L, Turesky RJ, Gremaud E, Trudel LJ, Stadler RH (1995) Determination of 8-oxoguanine in DNA by gas chromatography-mass spectrometry and HPLC-electrochemical detection: overestimation of the background level of the oxidized base by the gas chromatography-mass spectrometry assay. Chem Res Toxicol 8:1039–1045.

    Google Scholar 

  133. Hamberg M, Zhang L-Y (1995) Quantitative determination of 8-hydroxyguanine and guanine by isotope dilution mass spectrometry. Anal Biochem 229:336–344.

    Google Scholar 

  134. Douki T, Delatour T, Bianchini F, Cadet J (1996b) Observation and prevention of an artefactual formation of oxidized DNA bases and nucleosides in the GC-EIMS method. Carcinogenesis 17:347–353.

    Google Scholar 

  135. Cadet J, Douki T, Ravanat J-L (1997) Artifacts associated with the measurement of oxidized DNA bases. Environ Health Perspect 105:1034–1039.

    Google Scholar 

  136. Jaruga P, Speina E, Gackowski D, Tudek B, Olinski R (2000) Endogenous oxidative DNA base modifications analysed with repair enzymes and GC/MS technique. Nucleic Acids Res 28:E16.

    Google Scholar 

  137. Douki T, Martini R, Ravanat, J-L, Tureski RJ, Cadet J (1997) Measurement of 2,6-diamino-4-hydroxy-5-formamidopyrimidine and 8-oxo-7,8-dihydroguanine in isolated DNA exposed to gamma radiation in aqueous solution. Carcinogenesis 18:2385–2391.

    Google Scholar 

  138. Ravanat J-L, Douki T, Duez P, Gremaud E, Herbert K, Hofer T, Lassere L, Saint-Pierre C, Favier A, Cadet J (2002) Cellular background of 8-oxo-7,8-dihydro-2′-deoxyguanosine: an isotope based method to evaluate artefactual oxidation of DNA during its extraction and subsequent work-up. Carcinogenesis 23:1911–1918.

    Google Scholar 

  139. Chao MR, Yen CC, Hu CW (2008) Prevention of artifactual oxidation in determination of cellular 8-oxo-7,8-dihydro-2′-deoxyguanosine by isotope-dilution LC-MS/MS with automated solid-phase extraction. Free Radic Biol Med 44:464–473.

    Google Scholar 

  140. Mangal D, Vudathala D, Park J-H, Lee S H, Penning TM, Blair IA (2009) Analysis of 7,8-dihydro-8-oxo-2′-deoxyguanosine in cellular DNA during oxidative stress. Chem Res Toxicol 22:788–797.

    Google Scholar 

  141. Badouard C, Ménézo Y, Panteix G, Ravanat J-L, Douki T, Favier A (2008) Determination of new types of DNA lesions in human sperm. Zygote 16:9–13.

    Google Scholar 

  142. Collins AR, Cadet J, Möller L, Poulsen H, Viňa J (2004) Are we sure we know how to measure 8-oxo-7,8-dihydroguanine in DNA from human cells? Arch. Biochem. Biophys 423:57–65.

    Google Scholar 

  143. Gedik CM, Collins AR and ESCODD (2005) Establishing the background level of base oxidation in human lymphocyte DNA: results of an inter-laboratory validation study. FASEB J 19:82–84.

    Google Scholar 

  144. Ravanat J-L, Duretz B, Guiller A, Douki T, Cadet J (1998) Isotope dilution high-performance liquid chromatography-electrospray tandem mass spectrometry assay for the measurement of 8-oxo-7,8-dihydro-2′-deoxyguanosine in biological samples. J Chromatogr. B 715:349–356.

    Google Scholar 

  145. Glintborg B, Weimann A, Kensler TW, Poulsen HE (2006) Oltipraz chemoprevention trial in Qidong, People’s Republic of China:unaltered oxidative biomarkers. Free Radic Biol Med 41:1010–1014.

    Google Scholar 

  146. Singh R, Teichert F, Verschoyle RD, Kaur B, Vives M, Sharma RA, Steward WP, Gescher AJ, Farmer PB (2009) Simultaneous determination of 8-oxo-2′-deoxyguanosine and 8-oxo-2′-deoxyadenosine in DNA using online column-switching liquid chromatography/tandem mass spectrometry. Rapid Commun. Mass Spectrom 23:151–160.

    Google Scholar 

  147. Tuo J, Jaruga P, Rodriguez H, Bohr VA, Dizdaroglu M (2003a) Primary and 8-hydroxyadenine resulting from oxidative stress. FASEB J 17:668–674.

    Google Scholar 

  148. Tuo J, Jaruga P, Rodriguez H, Dizdaroglu M, Bohr VA (2003b) The cockayne syndrome group B gene product is involved in cellular repair of 8-hydroxyadenine in DNA. J Biol Chem 277:30832–30837.

    Google Scholar 

  149. Hailer MK, Slad PG, Martin BD, Sugden KD (2005) Nei deficient Escherichia coli are sensitive to chromate and accumulate the oxidized guanine lesion spiroiminodihydantoin. Chem Res Toxicol 18:1378–1383.

    Google Scholar 

  150. Collins AR, Duthie SJ Dobson VL (1993) Direct enzymic degradation of endogenous oxidative base damage in human lymphocyte DNA. Carcinogenesis 14:1733–1735.

    Google Scholar 

  151. Collins AR, Oscoz AA, Brunborg G, Gaivão I, Giovannelli L, Kruszewski M, Smith CC, Stetina R (2008) The comet assay: topical issues. Mutagenesis 23:143–151.

    Google Scholar 

  152. Pflaum M, Will O, Epe B (1997) Determination of steady-state levels of oxidative DNA base modifications in mammalian cells by means of repair endonucleases. Carcinogenesis 18:2225–2231.

    Google Scholar 

  153. Pflaum M, Will O, Malher HC, Epe B (1998) DNA oxidation products determined with repair endonucleases in mammalian cells: types, basal levels and influence of cell proliferation. Free Radic Res 29:585–598.

    Google Scholar 

  154. Hartwig A, Dally H, Schlepegrell R (1996) Sensitive analysis of DNA damage in mammalian cells: use of the bacterial Fpg protein in combination with alkaline unwinding. Toxicol Lett 88:85–90.

    Google Scholar 

  155. Moreno-Villanueva M, Ffeiffer R, Sindinger T, Leake A, Müller M, Kirkwood TB, Bürkle A (2009) A modified and automated version of the ‘Fluorimetric Detection of Alkaline DNA Unwinding’ method to quantify formation and repair of DNA strand breaks. BMC Biotechnol 9:39 (9 pages).

    Google Scholar 

  156. Smith CC, O’Donovan MR, Martin EA (2006) hOGG1 recognizes oxidative damage using the comet assay with greater specificity than FPG or ENDOIII. Mutagenesis 21:185–190.

    Google Scholar 

  157. ESCODD (2003) Measurement of DNA oxidation in human cells by chromatographic and enzymic methods. Free Radic Biol Med 34:1089–1099.

    Google Scholar 

  158. Loft S, Högh D, Mikkelsen L, Risom L, Forochhammer L, Möller P (2008) Biomarkers of oxidative damage to DNA and repair. Biochem Soc Trans 36:1071–1076.

    Google Scholar 

  159. Azqueta A, Shaposhnikov S, Collins AR (2009) DNA oxidation: Investigating its key role in environmental mutagenesis with the comet assay. Mutat Res 674:101–108.

    Google Scholar 

  160. Lorenzo Y, Azqueta A, Luna L, Bonilla F, Domínguez G, Collins AR (2009) The carotonoid beta-cryptoxanthin stimulates the repair of DNA oxidation damage in addition to acting as an antioxidant in human cells. Carcinogenesis 30:308–314.

    Google Scholar 

  161. Hackenberg S, Friehs G, Froelich K, Ginzkey C, Koehler C, Scherzed A, Burghartz M, Hagen R, Kleinsasser N (2010) Intracellular distribution, geno- and cytotoxic effects of nanosized titanium dioxide particles in the anatase crystal phase on human nasal mucosa cells. Toxicol Letter 19:9–14.

    Google Scholar 

  162. Szaflik JP, Rusin P, Zaleska-Zmijewska A, Kowalski M, Majsterek I, Szaflik J (2010) Reactive oxygen species promote localized DNA damage in glaucoma-iris tissues of elderly patients vulnerable to diabetic injury. Mutat Res 697:19–23.

    Google Scholar 

  163. Trapp C, McCullough AK, Epe B (2007) The basal levels of 8-oxoG and other oxidative modifications in intact mitochondrial DNA are low even in repair-deficient (Ogg1-/-/Csb-/-)mice Mutat Res 625:155–163.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jean Cadet .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2011 Springer Science+Business Media, LLC

About this chapter

Cite this chapter

Cadet, J., Douki, T., Ravanat, JL. (2011). Oxidatively Generated Damage to DNA and Biomarkers. In: Basu, S., Wiklund, L. (eds) Studies on Experimental Models. Oxidative Stress in Applied Basic Research and Clinical Practice. Humana Press, Totowa, NJ. https://doi.org/10.1007/978-1-60761-956-7_29

Download citation

Publish with us

Policies and ethics