DOI QR코드

DOI QR Code

A Critical Evaluation of DNA Adducts as Biological Markers for Human Exposure to Polycyclic Aromatic Compounds

  • Godschalk, Roger W.L. (Department of Health Risk Analysis and Toxicology, University of Maastricht) ;
  • Van Schooten, Frederik-Jan (Department of Health Risk Analysis and Toxicology, University of Maastricht) ;
  • Bartsch, Helmut (Division of Toxicology and Cancer Risk Factors, German Cancer Research Center)
  • Published : 2003.01.31

Abstract

The causative role of polycyclic aromatic hydrocarbons (PAH) in human carcinogenesis is undisputed. Measurements of PAH-DNA adduct levels in easily accessible white blood cells therefore represent useful early endpoints in exposure intervention of chemoprevention studies. The successful applicability of DNA adducts as early endpoints depends on several criteria:i.adduct levels in easily accessible surrogate tissues should reflect adduct levels in target-tissues, ii. toxicokinetics and the temporal relevance should be properly defined.iii. sources of inter- and intra-individual variability must be known and controllable, and finally iv. adduct analyses must have advantages as compared to other markers of PAH-exposure. In general, higher DNA adduct levels or a higher proportion of subjects with detectable DNA adduct levels were found in exposed individuals as compared with non-exposed subjects, but saturation may occur at high exposures. Furthermore, DNA adduct levels varied according to changes in exposure, for example smoking cessation resulted in lower DNA adduct levels and adduct levels paralleled seasonal variations of air-pollution. Intra-individual variation during continuous exposure was low over a short period of time (weeks), but varied significantly when longer time periods (months) were investigated. Inter-individual variation is currently only partly explained by genetic polymorphisms in genes involved in PAH-metabolism and deserves further investigation. DNA adduct measurement may have three advantages over traditional exposure assessment: i. they can smooth the extreme variability in exposure which is typical for environmental toxicants and may integrate exposure over a longer period of time. Therefore, DNA adduct assessment may reduce the monitoring effort. ii. Biological monitoring of DNA adducts accounts for all exposure routes. iii. DNA adducts may account for inter-individual differences in uptake, elimination, distribution, metabolism and repair amongst exposed individuals. In conclusion, there is now a sufficiently large scientific basis to justify the application of DNA adduct measurement as biomarkers in exposure assessment and intervention studies. Their use in risk-assessment, however, requires further investigation.

Keywords

References

  1. Alexandrov, K., Rojas, M., Geneste, O., Castegnaro, M., Camus, A.M., Petruzelli, S., Giuntini, C. and Bartsch, H. (1992) An improved fluoremetric assay for dosimetry of benzo$\left[\alpha \right]$pyrene diol-epoxide DNA adducts in smoker's lung: comparison with total bulky adducts and aryl hydrocarbon hydroxylase activity. Cancer Res. 52, 6248-6253.
  2. Alexandrov, K., Cascorbi, I., Rojas, M., Bouvier, G., Kriek, E. and Bartsch, H. (2002) CYPIAI and GSTMl genotypes affect benzo$\left(\alpha \right)$pyrene DNA adducts in smokers lung: comparison with aromatic/hydrophobic adduct formation. Carcinogenesis, 23, 1969-1977 https://doi.org/10.1093/carcin/23.12.1969
  3. Besarati Nia, A., Maa, L. M., Brouwer, E. M., Kleinjans, J. C. and Van Schooten, E J. (2000) Comparison between smoking- related DNA adduct analysis in induced sputum and peripheral blood lymphocytes. Carcinogenesis 21, 1335-1340. https://doi.org/10.1093/carcin/21.7.1335
  4. Burchell, B. and Coughtrie, M. W. (1997) Genetic and environmental factors associated with variation of human xenobiotic glucuronidation and sulfation. Environ. Health. Perspect. 105 Suppl., 739-747. https://doi.org/10.2307/3433277
  5. Cuzick, J., Routledge, M. N., Jenkins, D. and Gamer, R. C. (1990) DNA adducts in different tissues of smokers and non-smokers. Int. J. Cancer 45, 673-678. https://doi.org/10.1002/ijc.2910450417
  6. Dallinga, J. W., Pachen, D. M., Wijnhoven, S. W., Breedijk, A., van't Veer, L., Wigbout, G., van Zandwijk, N., Maas, L. M., van Agen, E., Kleinjans. J. C. and van Schooten, E J. (1998) The use of 4-aminobiphenyl hemoglobin adducts and aromatic DNA adducts in lymphocytes of smokers as biomarkers of exposure. Cancer Epidemiol. Biomarkers & PreV. 7, 571-577.
  7. De Flora, S., Izotti, A., D' Agostini. E., Rossi, G. A. and Balansky, R. M. (1993) Pulmonary alveolar macrophages in molecular epidemiology and chemoprevention of cancer. Environ. Health. Perspect. 99, 249-252. https://doi.org/10.2307/3431493
  8. Denissenko, M. E, Pao, A., Tang, M. and Pfeifer, G. P. (1996) Preferential formation of benzo$\left[\alpha \right]$pyrene adducts at lung cancer mutational hotspots in P53. Science 18, 430-432
  9. Denissenko, M. E, Pao, A., Pfeifer, G. P. and Tang, M. (1998) Slow repair of bulky DNA adducts along the nontranscribed strand of the human p53 gene may explain the strand bias of transversion mutations in cancer. Oncogene 16, 1241-1247. https://doi.org/10.1038/sj.onc.1201647
  10. D'Errico, Taioli, E., Chen, X. and Vineis. P. (1996) genetic metabolic polymorphisms and the risk of cancer: A review of the literature. Biomarkers 1, 149-173. https://doi.org/10.3109/13547509609079352
  11. Dickey. C., Santella, R. M., Hattis, D., Tang, D., Hsu, Y., Cooper, T., Young, T. L. and Perera, F. P. (1997) Variability in PAH-DNA adduct measurements in peripheral mononuclear cells: implications for quantitative cancer risk assessment. Risk Analysis 17, 649-656. https://doi.org/10.1111/j.1539-6924.1997.tb00905.x
  12. Dunn, B. P., Vedal, S., San, R. H. C., Kwan, W. F., Nelems, B., Enarson, D. A. and Stich, H. (1991) DNA adducts in bronchial biopsies. Int. J. Cancer 48. 485-492. https://doi.org/10.1002/ijc.2910480403
  13. Gamer, R. C., Cuzick, J., Jenkins, D., Phillips, D. H., Hewer, A., King, M. M. and Routledge, M. N. (1990) Linear relationship between DNA adducts in human lung and cigarette smoking. IARC Sci. Publ. 421-426.
  14. Godschalk, R. W. L., Vermeer, I. T. M., Kriek, E.. Floot, B., Schilderman, P. A. E. L., Moonen, E. J. C., Kleinjans, J. C. S. and Van Schooten, F. J. (1997) Comparison of $32_{P}$-postlabeling and HPLC-FD analysis of DNA adducts in rats acutely exposed to benzo$\left[\alpha \right]$pyrene. Chem. BioI. Inter. 104, 41-54. https://doi.org/10.1016/S0009-2797(97)03765-4
  15. Godschalk, R. W. L., Ostertag, J. U .. Moonen, E. J. C.. Neumann, H. A. M., Kleinjans, J. C. S. and Van Schooten, E J. (l998a) Aromatic DNA adducts in human white blood cells and skin after dermal application of coal-tat Cancer Epidemiol. Biomarkers & Prevo 7, 767-773.
  16. Godschalk, R. W. L., Maas, L. M., Van zandwijk, N., Van't veer, L., Breedijk, A., Borm, P. J. A., Verhaert, J., Kleinjans, J. C. S. and Van Schooten, F. J. (1998b) Differences in aromatic-DNA adduct levels between alveolar macrophages and subpopulations of white blood cells from smokers. Carcinogenesis 19, 819-825. https://doi.org/10.1093/carcin/19.5.819
  17. Godschalk. R. W. L.. Maas, L. M., Kleinjans, J. C. and Van Schooten, F. J. (1998c) Influences of DNA isolation and RNA contamination on carcinogen-DNA adduct analysis by $32_{P}$- postlabeling. Environ. Mol. Mutag. 32, 344-350. https://doi.org/10.1002/(SICI)1098-2280(1998)32:4<344::AID-EM8>3.0.CO;2-P
  18. Godschalk, R. W. L., Moonen, E. J., Schilderman, P. A., Broekmans, W. M., Kleinjans, J. C. and Van Schooten, F. J. (2000) Exposure route dependent formation of DNA adducts by polycyclic aromatic hydrocarbons. Carcinogenesis 21, 87- 92. https://doi.org/10.1093/carcin/21.1.87
  19. Godschalk, R. W. L., Dallinga, J. W., Wikman, H., Risch, A., Kleinjans, J. C., Bartsch, H. and Van Schooten, F. J. (2001) Modulation of DNA and protein adducts in smokers by genetic polymorphisms in GSTM1, GSTT1, NAT1 and NAT2. Pharmacogenetics 11, 389-398. https://doi.org/10.1097/00008571-200107000-00003
  20. Godschalk, R. W. L., Feldker, D. E., Borm, P. J., Wouters, E. F. and van Schooten, F. J. (2002) Body mass index modulates aromatic-DNA adduct levels and their persistence in smokers. Cancer Epidemiol. Biomarkers & Prev. 11, 790-793.
  21. Grzybowska, E., Hemminki, K., Szeliga, J. and Chorazy, M. (1993) Seasonal variation of aromatic DNA adducts in human lymphocytes and granulocytes. Carcinogenesis 14, 2523-2526. https://doi.org/10.1093/carcin/14.12.2523
  22. Gupta, R. C., Earley, K and Sharma, S. (1988) Use of human lymphocytes to measure DNA binding capacity of chemical carcinogens. Proc. Natl. Acad. Sci. 85, 3513-3517. https://doi.org/10.1073/pnas.85.10.3513
  23. Hall, M. and Grover, P. L. (1990) Polycyclic aromatic hydrocarbons: metabolism, activation and tumour initiation; in: Chemical Carcinogenesis and Mutagenesis, Cooper, C. S. and Grover, P. L. (eds.), Vol. I. pp. 327-372, Raven Press, New York, New York.
  24. Harris, R E., Zang, E. A, Anderson, J. I. and Wynder, E. L. (1993) Race and sex differences in lung cancer risk associated with cigarette smoking. Int. J. Epidemiol. 22, 592-599. https://doi.org/10.1093/ije/22.4.592
  25. Hawke, L. J. and Farrell, G. C. (1986) Increased binding of benzo(a)pyrene metabolites to lymphocytes from patients with lung cancer. Cancer Lett. 30, 289-297. https://doi.org/10.1016/0304-3835(86)90053-4
  26. Hayashi, S. I., Watanabe, J. and Nakachi, K (1991) Genetic linkage of lung cancer associated MspI polymorphisms with amino acid replacement in the heme binding region of the human cytochrome P4501A1 gene. J. Biochem. 110,407-411 https://doi.org/10.1093/oxfordjournals.jbchem.a123594
  27. Hemminki, K. (1993) DNA adducts, mutations and cancer. Carcinogenesis 14, 2007-2012. https://doi.org/10.1093/carcin/14.10.2007
  28. Herbert, R, Marcus, M., Wolff, M. S., Perera, F. P., Andrews, L., Godbold, J. H., Rivera, M., Stefanidis, M., Lu, X-Q., Landrigan, P. J. and Santella, R. M. (1990) Detection of adducts of deoxyribonucleic acid in white blood cells of roofers by $32_{P}$-postlabeling. Relationship of adduct levels to measures of polycyclic aromatic hydrocarbons. Scand. J. Work Environ. Health 16, 135-143. https://doi.org/10.5271/sjweh.1806
  29. Huang, M. -T., Wang, Z. Y., Georgiadis, C. A, Laskin, J. D. and Conney, A H. (1992) Inhibitory effects of curcumin on tumor initiation by benzo[a]pyrene and 7,12-dimethylbenz$\left[\alpha \right]$ anthracene. Carcinogenesis 13, 2183-2186.
  30. International Agency for research on Cancer (1983) Polynuclear Aromatic Compounds. Chemical. Environment and Experimental Data, IARC Monographs. Vol. 32. Lyon, France, International Agency for Research on cancer.
  31. Izzotti, A., Rossi, G. A. Bagnasco, M., de Flora S. (1991) Benzo[a]pyrene diolepoxide-DNA adducts in alveolar macrophages of smokers. Carcinogenesis 12, 1281- 1285. https://doi.org/10.1093/carcin/12.7.1281
  32. Jongeneelen, F. J., Anzion, R B. M., Scheepers, P. T. J., Bos, R P., Henderson, P. T., Nijenhuis, E. H., Veenstra, S. J., Brouns, R M. E. and Winkes, A. (1988) 1-Hydroxypyrene in urine as a biological indicator of exposure to polycyclic aromatic hydrocarbons in several work environments. Ann. Occup. Hygiene, 32, 35-43. https://doi.org/10.1093/annhyg/32.1.35
  33. Kang, D. H., Rothman, N., Poirier, M. C., Greenberg, A., Hsu, C. H., Schwartz, B. S., Baser, M. E., Weston, A., Groopman, J. D. and Strickland, P. T. (1995) Interindividual differences in the concentration of 1-hydroxypyrene-glucuronide in urine and polycyclic aromatic hydrocarbon-DNA adducts in peripheral white blood cells after charbroiled beef consumption. Carcinogenesis 16, 1079-1085. https://doi.org/10.1093/carcin/16.5.1079
  34. Kato, S., Bowman, E. D., Harrington, A. M., Blomeke, B. and Shields, P. (1995) Human lung carcinogen-DNA adduct levels mediated by genetic polymorphisms in vivo. J. Natl. Cancer Inst. 87, 902-907. https://doi.org/10.1093/jnci/87.12.902
  35. Kawajiri, K, Nakashi, K, Imai, K, Yoshii, A., Shinoda, N. and Watanabe, J. (1990) Identification of genetically high risk individuals to lung cancer by DNA polymorphisms of the cytochrome P4501A1 gene. FEBS Lett. 263, 131-133. https://doi.org/10.1016/0014-5793(90)80721-T
  36. Ketterer, B., Harris, J. M., Talaska, G., Meyer, D. J., Pemble, S. E., Taylor, J. B., Lang, N. P. and Kadlubar, F. F. (1992) The human glutathion S-transferase supergene family, its polymorphism and its effects on susceptibility to lung cancer. Environ. Health Perspect. 98, 87-94. https://doi.org/10.2307/3431252
  37. Knudsen, L. E., Ryder, L. P. and Wasserman, K (1992) induction of DNA repair synthesis in human monocytes/B-lymphocytes compared with T-Iymphocytes after exposure to N-acetoxy-N-acetylaminofluorene and dimethylsulfate in vitro. Carcinogenesis 13, 1285- 1287. https://doi.org/10.1093/carcin/13.7.1285
  38. Lewtas, J., Walsh, D., Williams, R. and Dobias, L. (1997) Air pollution exposure-DNA adduct dosimetry in humans and rodents: evidence for non-linearity at high doses. Mutat. Res. 378, 51-63. https://doi.org/10.1016/S0027-5107(97)00097-3
  39. Lutz, W. K. (1990) Dose-response relationship and low dose extrapolation in chemical carcinogenesis. Carcinogenesis 11, 1243-1247. https://doi.org/10.1093/carcin/11.8.1243
  40. Marshall, C. J., Vousden, K. H. and Phillips, D. H. (1984) Activation of c-H-ras-1 proto-oncogene by in vitro modification with a chemical carcinogen, benzo[a]pyrene-diol epoxide. Nature 310, 586-589. https://doi.org/10.1038/310586a0
  41. Mooney, L. V A, Santella, R M., Covey, L., Jeffrey, A. M., Bigbee, W., Randall, M. C., Cooper, T. B., Ottman, R, Tsai, W. Y., Wazneh, L., Glassman, A. H., Young, T. L. and Perera, F. P. (1995) Decline of DNA damage and other biomarkers in peripheral blood following smoking cessation. Cancer Epidemiol. Biomarkers & Prevo 4, 627-634.
  42. Nakachi, K, Imai, K., Hayashi, S. and Kawajiri, K (1993) Polymorphisms of the CYP1A1 and glutathione S-transferase genes associated with susceptibility to lung cancer in relation to cigartte dose in a Japanese population. Cancer Res. 53, 2994-2999.
  43. Nowak, D., Schmidt-Preuss, U., Jorres, R, Liebke, F. and RUdiger, H. W. (1988) Formation of adducts and water-soluble metabolites of B(a)P in human monocytes is genetically controled. Int. J. Cancer 41, 169-173. https://doi.org/10.1002/ijc.2910410202
  44. Nowak, D., Meyer, A, Schmidt-Preuss, U., Gatzemeier, U., Magnussen, H. and Rudiger. H. W. (1992) Formation of benzo(a)pyrene-DNA adducts in blood monocytes from lung cancer patients with a familial history of lung cancer. J. Cancer Res. Clin. Oneol. 118, 67-71. https://doi.org/10.1007/BF01192314
  45. Okano, P., Miller, H. N., Robinson, R. C. and Gelboin, H. V. (1979) Comparison of benzo[a]pyrene and (-)-trans-7,8- dihydroxy-7,8-dihydrobenzo[aJpyrene metabolism in human blood monocytes and lymphocytes. Cancer Res. 39,3184-3193.
  46. Paleologo, M., van Schooten, F. J., Pavanello, S., Kriek, E., Zordan, M., Clonfero, E., Bezze, C. and Levis, A G. (1992) Detection of benzo(a)pyrene-diol-epoxide-DNA adducts in white blood cells of psoriatic patients treated with coal tar. Murat. Res. 281, 11-16. https://doi.org/10.1016/0165-7992(92)90030-L
  47. Palli, D., Vineis, P., Russo, A., Berrino, E, Krogh, V., Masala, G., Munnia, A., Panico, S., Taioli, E., Tumino, R, Garte, S. and Peluso, M. (2000) Diet, metabolic polymorphisms and DNA adducts: the EPIC-Italy cross-sectional study. Int J Cancer. 87, 44-451. https://doi.org/10.1002/1097-0215(20000701)87:1<44::AID-IJC7>3.0.CO;2-Q
  48. Pavord, I. D., Pizzichini, M. M. M., Pizzichini, E. and Hargreave, F. E. (1997) The use of induced sputum to investigate airway inflammation. Thorax 52, 498-501. https://doi.org/10.1136/thx.52.6.498
  49. Peluso, M., Ceppi, M., Munnia, A., Puntoni, R and Parodi, S. (2001) Analysis of 13 $32_{P}$-postlabeling studies on occupational Cohorts exposed to air pollution. Am. J. Epidemiol. 153, 546-558. https://doi.org/10.1093/aje/153.6.546
  50. Perera, F. P., Mayer, J., Jaretzki, A., heamc, S., brenner, D., Young, T. L., Fischman, H. K., Grimes, M., Grantham, S. and Tang, M. X. (1989) Comparison of DNA adducts and sister chromatid exchange in lung cancer cases and controls. Cancer Res. 49, 4446-4451.
  51. Perera, F. P., Dickey, C., santella, R, O'Neill, J. P., Albertini, R. J., Ottman, R, Tsai. W. Y., Mooney, L. A., Savela, K. and Hemminki, K. (1994) Carcinogen-DNA adducts and genemutations in foundry workers with low-level exposure to polycyclic aromatic hydrocarbons. Carcinogenesis 15 2905-2910. https://doi.org/10.1093/carcin/15.12.2905
  52. Phillips, D. H., Hewer, A. and Grover, P. L. (1986) Aromatic DNA adducts in human bone marrow and peripheral blood leukocytes. Carcinogenesis 7, 2071-2075. https://doi.org/10.1093/carcin/7.12.2071
  53. Phillips, D. H., Hewer, A., Martin, C. N., Gamer, R C. and King, M. M. (1988) Correlation of DNA adduct levels in human lung with cigarette smoking. Nature 336, 790-792. https://doi.org/10.1038/336790a0
  54. Phillips, D. H .. Schoket, B., Hewer, A., Bailey, E., Kostic, S. and Vincze, l. (1990) Influence of cigarette smoking on the levels of DNA adducts in human bronchial epithelium and white blood cells. Int. J. Cancer 46, 569-575. https://doi.org/10.1002/ijc.2910460403
  55. Phillips, D. H. and Castegnaro, M. (1999) Standardization and validation of DNA adduct postlabeling methods: report of interlaboratory trials and production of recommended protocols. Mutagenesis 14, 301-315. https://doi.org/10.1093/mutage/14.3.301
  56. Poirier, M. and Beland, E A. (1992) DNA adduct measurements and tumour incidence during chronic carcinogen exposure in animal models:implications for DNA adduct based human cancer risk assesment, Chem. Res. Toxicol. 5, 749-755. https://doi.org/10.1021/tx00030a003
  57. Risch, H. A., Howe, G. R, Jain, M., Burch, J. D., Holoway, E. J. and Miller, A. B. (1993) Are female smokers at higher risk for lung cancer than male smokers? A case-control analysis by histological type. Am. J. Epidemiol. 138, 281-293. https://doi.org/10.1093/oxfordjournals.aje.a116857
  58. Rojas, M., Alexandrov, K., Cascorbi, l., Brockmoller, J., Likhachev, A., Pozharisski, K., Bouvier, G., Auburtin, G., Mayer, L., Kopp-Schneider, A., Roots, I. and Bartsch, H. (1998) High Beno[a]pyrene diol-epoxide DNA adduct levels in lung and blood cells from individuals with combined CYP1A1 Mspl/lMspl-GSTM1$*_{0}/*_{0}$ genotypes. Phannacogenet. 8. 109-118.
  59. Ross, J. A., Nelson, G., Kligerman, A., Erexson, G., Bryant, M., Earley, K., Gupta, R. and Nesnow, S. (1990) Formation and persistence of novel B(a)P adducts in rat lung, liver, and peripheral blood lymphocyte DNA. Cancer Res. 50, 5088-5094.
  60. Ross, J. A., Nelson, G. B., Wilson, K. H., Rabinowitz, J. R, Galati, A, Stoner, G. D., Nesnow, S., Mass, M. J. (1995) Adenomas induced by polycyclic aromatic hydrocarbons in strain A/J mouse lung correlate with time-integrated DNA adduct levels. Cancer Res. 55, 1039-1044.
  61. Rothman, N., Correa-Villasenor, A., Ford, D. P., Poirier, M. C., Haas, R. A., Hansen, J. A., O'Toole, T. and Strickland, P. T. (1993a) Contribution of occupation and diet to white blood cell PAR DNA adducts in wildland firefighters. Cancer Epidemiol. Biomarkers & Prevo 2, 341-347.
  62. Rothman, N., Poirier, M. C., Haas, R A, Correa-Villasenor, A., Ford, P., Hansen, J. A., O'Toole, T. and Strickland, P. T. (1993b) Association of PAH DNA adducts in peripheral white blood cells with dietary exposure to PAH. Environ. Health Perspect. 99, 265-267. https://doi.org/10.2307/3431497
  63. Ryberg, D., Hewer, A., Phillips, D. H. and Haugen A. (1994) Different susceptibility to smoking-induced DNA damage among male and female lung cancer patients. Cancer Res. 54, 5801-5803.
  64. Santella, R.M., Hemminki, K., Tang, D. L., Paik, M., Ottman, R., Young, T. L., Savela, K., Vodickova, L., Dickey, C., Whyatt, R. and Perera, F. P. (1993) Polycyclic aromatic hydrocarbon- DNA adducts in white blood cells and urinary 1-hydroxypyrene in foundry workers. Cancer Epidemiol. Biomarkers & Prev. 2, 59- 62.
  65. Schoket, B., Kostic, S. and Vince, l. (1993) determination of smoking-related DNA adducts in lung-cancer and non-cancer patients. IARC Sci. Publ. 315-319.
  66. Schoket, B., Phillips, D. H., Kostic, S. and Vincze, l. (1998) Smoking associated bulky DNA adducts in bronchial tissue related to CYPJ1A1 Mspl and GSTM1 genotypes in lung patients. Carcinogenesis 19, 841-856. https://doi.org/10.1093/carcin/19.5.841
  67. Schulte, P. A. and Perera, F. P. (1993) Molecular Epidemiology. Principles and Practices, Academic Press, San Diego, California.
  68. Schut, H. A. J. and Shiverick, K. T. (1992) DNA adducts in humans as dosimeters of exposure to environmental, occupational or dietary genotoxins. FASEB J. 6, 2942-2951. https://doi.org/10.1096/fasebj.6.11.1644258
  69. Shields, P. G., Bowman, E. D., Harrington, A. M., Doan, V. T. and Weston, A. (1993) Polycyclic aromatic hydrocarbon-DNA adducts in human lung and cancer susceptibility genes. Cancer Res. 53, 3486-3492.
  70. Sugimura, H., Suzuki, I., Hamada, G. S., Iwase, T., Takahashi, T, Nagura, K., Iwata, H., Watanabe, S., Kino, I. and Tsugane, S. (1994) Cytochrome P450 1A1 genotype in lung cancer patients and controls in Rio de Janeiro, Brazil. Cancer Epidemiol. Biomarkers & Prev., 3, 145-148.
  71. Strickland, P., Kang. D. and Sithisarankul, P. (1996) Polycyclic aromatic hydrocarbon metabolites in urine as biomarkers of exposure and effect. Environ. Health Perspect. 104, Suppl. 5, 927-932. https://doi.org/10.2307/3433012
  72. Tang, D., Santella, R. M., Blackwood, A. M., Young, T. -L., Mayer, J .. Jaretzki, A., Grantham, S., Tsai, W.- Y. and Perera, F. P. (1995) A molecular epidemiological case-control study of lung cancer. Cancer Epidemiol. Biomarkers & Prev. 4, 341-346.
  73. Tang, D., Phillips, D. H., Stampfer, M., Mooney, L. A., Hsu, Y., Cho, S., Tsai. W. Y, Ma, J., Cole. K. J., She, M. N. and Perera, F. P. (2001) Association between carcinogen-DNA adducts in white blood cells and lung cancer risk in the physicians health study. Cancer Res. 61, 6708-6712.
  74. Tang, D., Cho, S., Rundle, A., Chen, S., Phillips, D., Zhou, J., Hsu, Y., Schnabel. F., Estabrook, A. and Perera, F. P. (2002) Polymorphisms in the DNA repair enzyme XPD are associated with increased levels of PAH-DNA adducts in a case-control study of breast cancer. Breast Cancer Res Treat. 75, 159-166. https://doi.org/10.1023/A:1019693504183
  75. Van Maanen, J. M. S., Maas. L. M., Hageman G., KIeinjans, J. C. S. and Van Agen, B. (l994a) DNA adduct and mutation analysis in white blood cells of smokers and non smokers. Environ. Mol. Mutag. 24, 46-50. https://doi.org/10.1002/em.2850240106
  76. Van Maanen. J. M. S., Moonen, E. J. C., Maas, L. M., KIeinjans, J. C. S., Van Schooten, F. J. (l994b) Formation of aromatic DNA adducts inwhite blood cells in relation to urinruy excretion of 1-hydroxy-pyrene during consumption of grilled meat. Carcinogenesis 15, 2263-2268. https://doi.org/10.1093/carcin/15.10.2263
  77. Van Schooten F. J., Hillebrand M. J. X., Scherer E, Den Engelse, L., Kriek, E. (1991) Immunocytochemical visualization of DNA adducts in mouse tissues and human white blood cells following treatment with B(a)P or its diol epoxide. A quantitative approach. Carcinogenesis 12, 427-433. https://doi.org/10.1093/carcin/12.3.427
  78. Van Schooten F. J., Hillebrand, M. J. X., Van Leeuwen, F. E., Van Zandwijk, N., Jansen, H. M., Den Engelse, L. and Kriek, E. (1992) Polycyclic aromatic hydrocarbon-?-DNA adducts in white blood ceIls from lung cancer patients: no correlation with adduct levels in lung. Carcinogenesis 13, 987-993. https://doi.org/10.1093/carcin/13.6.987
  79. Van Schooten, E J., Jongeneelen, E J., Hillebrand, M. J. X., Van leeuwen, F. E., de Looff, A J. A, Dijkmans. A P. G., van Rooij, J. G. M., den Engelse, L. and Kriek, E. (1995) Polycyclic aromatic hydrocarbon-DNA adducts in white blood cell DNA and 1-hydroxypyrene in the urine from aluminum workers: relation with job category and synergistic effects of smoking. Cancer Epidemiol. Biomarkers & Prev. 4, 69-77.
  80. Van Schooten. E J., Godschalk, R. W. L., Breedijk, A, Maas, L. M., Kriek, E., Sakai, H., Wigbout, G., Baas, P., Van't Veer. L. and Van Zandwijk, N. (1997) $32_{P}$-PostiabeIling of aromatic DNA adducts in white blood cells and alveolar macrophages of smokers: saturation at high exposures. Mutat. Res. 378, 65-75. https://doi.org/10.1016/S0027-5107(97)00098-5
  81. Van Schooten, F. J., Nia, A. B., De Flora, S., D' Agostini, F., Izzotti, A., Camoirano, A., Balm, A. J., Dallinga, J. W., Bast, A, Haenen. G. R., Vant Veer, L., Baas, P., Sakai, H. and Van Zandwijk. N. (2002) Effects of oral administration of N-acetyl- L-cysteine: a multi-biomarker study in smokers, Cancer Epidemiol. Biomarkers & Prev. 11, 167-75.
  82. Vineis, P. and Caporaso. N. (1995) Tobacco and cancer: Epidemiology and the laboratory. Environ. Health Perspect. 103, 156-160. https://doi.org/10.2307/3432271
  83. Vousden. K. H., Bos, J. L., Marshall, C. J., Phillips, D. H. (1986) Mutations activating human c-Ha-ras 1 protooncogene (HRAS1) induced by chemical carcinogens and depurination. Proc. Natl. Acad. Sci. USA 83, 1222-1226. https://doi.org/10.1073/pnas.83.5.1222
  84. Wieneke, J. K. Kelsey, K. T., Varkonyi, A., Semey, K, Wain, J. C., Mark, E., Christiani, D. C. (1995) Correlation of DNA adducts in blood mononuclear cells with tobacco carcinogen- induced damage in human lung. Cancer Res. 55, 4910 4914.
  85. Wogan, G. N. and Gorelick, N. J. (1985) Chemical and biochemical dosimetry of exposure to genotoxic chemicals. Environ. Health Perspect. 62, 5-18. https://doi.org/10.2307/3430088
  86. Xu, X., Kelsey, K. T., Wieneke, J. K, Wain, J. C. and Christiani, D. C. (1996) Cytochrome P4501A1 Mspl polymorphism and lung cancer susceptibility. Cancer Epidemiol. Biomarkers & Prev. 5, 687-692.
  87. Yang, J. -L., Maher, V. M. and McCormick, J. J. (1987) Kinds of mutations formed when a shuttle vector containing adducts of $\left({\pm} \right)$-7$\beta$,8$\alpha$-dihydroxy-9$\alpha$, 10$\alpha$-epoxy-7,8,9,10-tetrahydrobenzo- [a]pyrene replicates in human cells. Proc. Natl. Acad. Sci. USA 84. 3787-3791. https://doi.org/10.1073/pnas.84.11.3787

Cited by

  1. 1,10-Phenanthroline stabilizes mRNA of the carcinogen-metabolizing enzyme, cytochrome P450 1a1 vol.192, pp.2, 2010, https://doi.org/10.1016/j.toxlet.2009.10.029
  2. Polycyclic aromatic hydrocarbon (PAH)–DNA adducts and breast cancer: modification by gene promoter methylation in a population-based study vol.26, pp.12, 2015, https://doi.org/10.1007/s10552-015-0672-7
  3. Aromatic DNA adducts in relation to dietary and other lifestyle factors in Spanish adults vol.229, pp.4, 2009, https://doi.org/10.1007/s00217-009-1089-x
  4. Toxicogenomic outcomes predictive of forestomach carcinogenesis following exposure to benzo(a)pyrene: Relevance to human cancer risk vol.273, pp.2, 2013, https://doi.org/10.1016/j.taap.2013.05.027
  5. Aldo-keto reductase 1C2 is essential for 1-nitropyrene's but not for benzo[a]pyrene's induction of p53 phosphorylation and apoptosis vol.244, pp.2-3, 2008, https://doi.org/10.1016/j.tox.2007.11.022
  6. Hair analysis for biomonitoring of environmental and occupational exposure to organic pollutants: State of the art, critical review and future needs vol.210, pp.2, 2012, https://doi.org/10.1016/j.toxlet.2011.10.021
  7. Biomonitoring of complex occupational exposures to carcinogens: The case of sewage workers in Paris vol.8, pp.1, 2008, https://doi.org/10.1186/1471-2407-8-67
  8. Simultaneous Detection of Multiple DNA Adducts in Human Lung Samples by Isotope-Dilution UPLC-MS/MS vol.87, pp.1, 2015, https://doi.org/10.1021/ac503803m
  9. Air pollution by carcinogenic PAHs and plasma levels of p53 and p21WAF1 proteins vol.620, pp.1-2, 2007, https://doi.org/10.1016/j.mrfmmm.2007.02.020
  10. Monitoring of DNA Adducts in Humans and 32P-Postlabelling Methods. A Review vol.69, pp.3, 2004, https://doi.org/10.1135/cccc20040476
  11. Punicalagin and Ellagic Acid Demonstrate Antimutagenic Activity and Inhibition of Benzo[a]pyrene Induced DNA Adducts vol.2014, 2014, https://doi.org/10.1155/2014/467465
  12. Longitudinal effects of prenatal exposure to air pollutants on self-regulatory capacities and social competence vol.57, pp.7, 2016, https://doi.org/10.1111/jcpp.12548
  13. Development and validation of a direct sandwich chemiluminescence immunoassay for measuring DNA adducts of benzo[a]pyrene and other polycyclic aromatic hydrocarbons vol.27, pp.5, 2012, https://doi.org/10.1093/mutage/ges024
  14. A Review on the Practical Application of Human Biomonitoring in Integrated Environmental Health Impact Assessment vol.12, pp.2, 2009, https://doi.org/10.1080/15287390802706397
  15. Oral Bioavailability, Bioaccessibility, and Dermal Absorption of PAHs from Soil—State of the Science vol.50, pp.5, 2016, https://doi.org/10.1021/acs.est.5b04110
  16. DNA and protein adducts in human tissues resulting from exposure to tobacco smoke vol.131, pp.12, 2012, https://doi.org/10.1002/ijc.27827
  17. Albumin and hemoglobin adducts of benzo[a]pyrene in humans—Analytical methods, exposure assessment, and recommendations for future directions vol.40, pp.2, 2010, https://doi.org/10.3109/10408440903283633
  18. Genetic and epigenetic cancer chemoprevention on molecular targets during multistage carcinogenesis vol.90, pp.10, 2016, https://doi.org/10.1007/s00204-016-1813-9
  19. Prenatal exposure to polycyclic aromatic hydrocarbons and cognitive dysfunction in children vol.22, pp.5, 2015, https://doi.org/10.1007/s11356-014-3627-8
  20. 10.1002/0471435139.hyg082 vol.1, 2000, https://doi.org/10.1002/0471435139.hyg082
  21. Seasonal variations of DNA-adduct patterns in open field farmers handling pesticides vol.587, pp.1-2, 2005, https://doi.org/10.1016/j.mrgentox.2005.05.013
  22. Enhanced activity of punicalagin delivered via polymeric implants against benzo[a]pyrene-induced DNA adducts vol.743, pp.1-2, 2012, https://doi.org/10.1016/j.mrgentox.2011.12.022
  23. Comparative transcriptomic analyses to scrutinize the assumption that genotoxic PAHs exert effects via a common mode of action vol.90, pp.10, 2016, https://doi.org/10.1007/s00204-015-1595-5
  24. Validation of biomarkers for the study of environmental carcinogens: a review vol.13, pp.5, 2008, https://doi.org/10.1080/13547500802054611
  25. Biological monitoring of carcinogens: current status and perspectives vol.86, pp.4, 2012, https://doi.org/10.1007/s00204-011-0793-z
  26. Expression of XRCC5 in peripheral blood lymphocytes is upregulated in subjects from a heavily polluted region in the Czech Republic vol.713, pp.1-2, 2011, https://doi.org/10.1016/j.mrfmmm.2011.06.001
  27. PAH–DNA adducts in environmentally exposed population in relation to metabolic and DNA repair gene polymorphisms vol.620, pp.1-2, 2007, https://doi.org/10.1016/j.mrfmmm.2007.02.022
  28. F2RL3 Methylation as a Biomarker of Current and Lifetime Smoking Exposures 2013, https://doi.org/10.1289/ehp.1306937
  29. A sandwich ELISA for measuring benzo[a]pyrene–albumin adducts in human plasma vol.435, pp.2, 2013, https://doi.org/10.1016/j.ab.2012.12.021
  30. Quantified relations between exposure to tobacco smoking and bladder cancer risk: a meta-analysis of 89 observational studies vol.45, pp.3, 2016, https://doi.org/10.1093/ije/dyw044
  31. Aromatic DNA adducts and polymorphisms in metabolic genes in healthy adults: findings from the EPIC-Spain cohort vol.30, pp.6, 2009, https://doi.org/10.1093/carcin/bgp062
  32. Functional effect of polymorphisms in 15q25 locus on CHRNA5 mRNA, bulky DNA adducts andTP53mutations vol.132, pp.8, 2013, https://doi.org/10.1002/ijc.27870
  33. DNA methylation of the CYP1A1 enhancer is associated with smoking-induced genetic alterations in human lung vol.131, pp.7, 2012, https://doi.org/10.1002/ijc.27421
  34. Bioinformatics Analysis of the Effects of Tobacco Smoke on Gene Expression vol.10, pp.12, 2015, https://doi.org/10.1371/journal.pone.0143377
  35. Characterization of the Exposure-Disease Continuum in Neonates of Mothers Exposed to Carcinogens during Pregnancy vol.102, pp.2, 2008, https://doi.org/10.1111/j.1742-7843.2007.00174.x
  36. THE RELATION BETWEEN BIOMARKERS AND OCCUPATIONAL EXPOSURE TO POLYCYCLIC AROMATIC COMPOUNDS vol.24, pp.4-5, 2004, https://doi.org/10.1080/10406630490468748
  37. Structure, function and carcinogenicity of metabolites of methylated and non-methylated polycyclic aromatic hydrocarbons: a comprehensive review vol.26, pp.3, 2016, https://doi.org/10.3109/15376516.2015.1135223
  38. Chemistry and Structural Biology of DNA Damage and Biological Consequences vol.8, pp.9, 2011, https://doi.org/10.1002/cbdv.201100033
  39. Variation in PAH-related DNA adduct levels among non-smokers: The role of multiple genetic polymorphisms and nucleotide excision repair phenotype vol.132, pp.12, 2013, https://doi.org/10.1002/ijc.27953
  40. In vitro evaluation of baseline and induced DNA damage in human sperm exposed to benzo[a]pyrene or its metabolite benzo[a]pyrene-7,8-diol-9,10-epoxide, using the comet assay vol.25, pp.4, 2010, https://doi.org/10.1093/mutage/geq024
  41. Interaction of benzo[a]pyrene with other risk factors in hepatocellular carcinoma: a case-control study in Xiamen, China vol.24, pp.2, 2014, https://doi.org/10.1016/j.annepidem.2013.10.019
  42. Detection of benzo[a]pyrene-guanine adducts in single-stranded DNA using the α-hemolysin nanopore vol.26, pp.7, 2015, https://doi.org/10.1088/0957-4484/26/7/074002
  43. Factors and Trends Affecting the Identification of a Reliable Biomarker for Diesel Exhaust Exposure vol.44, pp.16, 2014, https://doi.org/10.1080/10643389.2013.790748
  44. The relationship between prenatal exposure to airborne polycyclic aromatic hydrocarbons (PAHs) and PAH–DNA adducts in cord blood vol.23, pp.4, 2013, https://doi.org/10.1038/jes.2012.117
  45. Effects of environmental air pollution on endogenous oxidative DNA damage in humans vol.620, pp.1-2, 2007, https://doi.org/10.1016/j.mrfmmm.2007.02.024
  46. Impact of barbecued meat consumed in pregnancy on birth outcomes accounting for personal prenatal exposure to airborne polycyclic aromatic hydrocarbons: Birth cohort study in Poland vol.28, pp.4, 2012, https://doi.org/10.1016/j.nut.2011.07.020
  47. Seasonal variability of oxidative stress markers in city bus drivers vol.642, pp.1-2, 2008, https://doi.org/10.1016/j.mrfmmm.2008.03.003
  48. Translational Pharmacokinetic-Pharmacodynamic Modeling from Nonclinical to Clinical Development: A Case Study of Anticancer Drug, Crizotinib vol.15, pp.2, 2013, https://doi.org/10.1208/s12248-012-9436-4
  49. Sensitivity of different endpoints for in vitro measurement of genotoxicity of extractable organic matter associated with ambient airborne particles (PM10) vol.620, pp.1-2, 2007, https://doi.org/10.1016/j.mrfmmm.2007.02.026
  50. Analysis ofN7-(benzo[a]pyrene-6-yl)guanine in urine using two-step solid-phase extraction and isotope dilution with liquid chromatography/tandem mass spectrometry vol.19, pp.7, 2005, https://doi.org/10.1002/rcm.1868
  51. Oncogene and tumor-suppressor gene products as serum biomarkers in occupational-derived lung cancer vol.7, pp.5, 2007, https://doi.org/10.1586/14737159.7.5.555
  52. Phytotherapeutic approach: a new hope for polycyclic aromatic hydrocarbons induced cellular disorders, autophagic and apoptotic cell death vol.27, pp.1, 2017, https://doi.org/10.1080/15376516.2016.1268228
  53. Polycyclic aromatic hydrocarbons and childhood asthma vol.30, pp.2, 2015, https://doi.org/10.1007/s10654-015-9988-6
  54. 10.1002/0471125326.hyg082 vol.1, 2000, https://doi.org/10.1002/0471125326.hyg082
  55. Impact of prenatal exposure to polycyclic aromatic hydrocarbons from maternal diet on birth outcomes: a birth cohort study in Korea vol.19, pp.14, 2016, https://doi.org/10.1017/S1368980016000550
  56. Human biomonitoring: State of the art vol.210, pp.3-4, 2007, https://doi.org/10.1016/j.ijheh.2007.01.024
  57. Fluoranthene enhances p53 expression and decreases mutagenesis induced by benzo[a]pyrene vol.208, pp.3, 2012, https://doi.org/10.1016/j.toxlet.2011.11.011
  58. Lifestyle, Environmental, and Genetic Predictors of Bulky DNA Adducts in a Study Population Nested within a Prospective Danish Cohort vol.73, pp.9, 2010, https://doi.org/10.1080/15287390903566633
  59. Developmental Brain and Behavior Toxicity of Air Pollutants: A Focus on the Effects of Polycyclic Aromatic Hydrocarbons (PAHs) vol.41, pp.22, 2011, https://doi.org/10.1080/10643389.2010.495644
  60. Polycyclic aromatic hydrocarbon- and aflatoxin-albumin adducts, hepatitis B virus infection and hepatocellular carcinoma in Taiwan vol.252, pp.1, 2007, https://doi.org/10.1016/j.canlet.2006.12.010
  61. Assessment of alteration of reproductive system in vivo induced by subchronic exposure to benzo(a)pyrene via oral administration vol.30, pp.1, 2015, https://doi.org/10.1002/tox.21889
  62. The role of reactive oxygen species and oxidative stress in environmental carcinogenesis and biomarker development vol.188, pp.2, 2010, https://doi.org/10.1016/j.cbi.2010.07.010
  63. Purple Rice Extract Supplemented Diet Reduces DMH-Induced Aberrant Crypt Foci in the Rat Colon by Inhibition of Bacterial β-Glucuronidase vol.15, pp.2, 2014, https://doi.org/10.7314/APJCP.2014.15.2.749
  64. Quantification of Hemoglobin and White Blood Cell DNA Adducts of the Tobacco Carcinogens 2-Amino-9H-pyrido[2,3-b]indole and 4-Aminobiphenyl Formed in Humans by Nanoflow Liquid Chromatography/Ion Trap Multistage Mass Spectrometry vol.30, pp.6, 2017, https://doi.org/10.1021/acs.chemrestox.7b00072
  65. Exposure assessment at the workplace: Implications of biological variability vol.168, pp.3, 2007, https://doi.org/10.1016/j.toxlet.2006.09.014
  66. cellular VOC excretion vol.12, pp.2, 2018, https://doi.org/10.1088/1752-7163/aa9080