DOI QR코드

DOI QR Code

The Role of Gene-environment Interaction in Environmental Carcinogenesis

환경성 발암 기전에서 유전자-환경 상호작용의 역할

  • Han, So-Hee (Department of Preventive Medicine, Seoul National University College of Medicine) ;
  • Lee, Kyoung-Mu (Department of Preventive Medicine, Seoul National University College of Medicine)
  • 한소희 (서울대학교 의과대학 예방의학교실) ;
  • 이경무 (서울대학교 의과대학 예방의학교실)
  • Received : 2010.02.01
  • Accepted : 2010.02.24
  • Published : 2010.02.28

Abstract

Evidences supporting gene-environment interaction are accumulating in terms of environmental exposure including lifestyle factors and related genetic variants. One form of defense mechanism against cancer development involves a series of genes whose role is to metabolize (activation/detoxification) and excrete potentially toxic compounds and to repair subtle mistakes in DNA. The purpose of this article is to provide a brief review of the notion of gene-environment interaction, environmental/occupational carcinogens and related cancers, and previous studies of gene-environment interaction on cancers caused by exposure to carcinogenesis. With a number of studies on the interaction between lifestyle factors (e.g., smoking and diet) and genetic polymorphisms in genes involved in xenobiotic metabolism and DNA repair excluded, only several studies have been conducted on the interactive effects between polymorphisms of CYPs, GSTs, ERCCs, XRCCs and environmental/occupational carcinogens such as vinyl chloride, benzo[a]pyrene, and chloroform on carcinogenesis or genotoxicity. Future studies may need to be conducted with sufficient number of subjects and based on occupational cohorts to provide useful information in terms of advanced risk assessment and regulation of exposure level.

Keywords

References

  1. Gordon, J. E. : Epidemiology in modern perspective. Proceedings of the Royal Society of Medicine, 47(7), 564-70, 1954.
  2. Hung, R. J., Boffetta, P., Brockmöller, J., Butkiewicz, D., Cascorbi, I., Clapper, M. L., Garte, S., Haugen, A., Hirvonen, A., Anttila, S, Kalina, I., Le Marchand, L., London, S. J., Rannug, A., Romkes, M., Salagovic, J., Schoket, B., Gaspari, L., Taioli, E. : CYP1A1 and GSTM1 genetic polymorphisms and lung cancer risk in Caucasian non-smokers: a pooled analysis. Carcinogenesis, 24(5), 875-882, 2003. https://doi.org/10.1093/carcin/bgg026
  3. Berwick, M., Vineis, P. : Markers of DNA repair and susceptibility to cancer in humans: an epidemiologic review. Journal of the National Cancer Institute, 92(11), 874-897, 2000. https://doi.org/10.1093/jnci/92.11.874
  4. Yang, C. X., Matsuo, K., Wang, Z. M., Tajima, K. : Phase I/II enzyme gene polymorphisms and esophageal cancer risk: a meta-analysis of the literature. World Journal of Gastroenterology, 11, 2531-2538, 2005. https://doi.org/10.3748/wjg.v11.i17.2531
  5. Lee, K. M., Kang, D., Clapper, M. L., Ingelman- Sundberg, M., Ono-Kihara, M., Kiyohara, C., Min, S., Lan, Q., Le Marchand, L., Lin, P., Lung, M. L., Pinarbasi, H., Pisani, P., Srivatanakul, P., Seow, A., Sugimura, H., Tokudome, S., Yokota, J., Taioli, E. : CYP1A1, GSTM1, and GSTT1 polymorphisms, smoking, and lung cancer risk in a pooled analysis among Asian populations. Cancer Epidemiology Biomarkers & Prevention, 17(5), 1120-1126, 2008. https://doi.org/10.1158/1055-9965.EPI-07-2786
  6. Shi, X., Zhou, S., Wang, Z., Zhou, Z. : CYP1A1 and GSTM1 polymorphisms and lung cancer risk in Chinese populations: a meta-analysis. Lung Cancer, 59, 155-163, 2008. https://doi.org/10.1016/j.lungcan.2007.08.004
  7. Hashibe, M., Brennan, P., Strange, R. C., Bhisey, R., Cascorbi, I., Lazarus, P., Oude Ophuis, M. B., Benhamou, S., Foulkes, W. D., Katoh, T., Coutelle, C., Romkes, M., Gaspari, L., Taioli, E., Boffetta, P. : Meta- and pooled analyses of GSTM1, GSTT1, GSTP1, and CYP1A1 genotypes and risk of head and neck cancer. Cancer Epidemiology, Biomarkers & Prevention, 12, 1509-1517, 2003.
  8. Zhuo, X., Cai, L., Xiang, Z., Li, Q., Zhang, X. : GSTM1 and GSTT1 polymorphisms and nasopharyngeal cancer risk: an evidence-based meta-analysis. Journal of Experimental & Clinical Cancer Research, 28(1), 46, 2009. https://doi.org/10.1186/1756-9966-28-46
  9. Capella, G., Pera, G., Sala, N., Agudo, A., Rico, F., Del Giudicce, G., Plebani, M., Palli, D., Boeing, H., Bueno-de-Mesquita, H. B., Carneiro, F., Berrino, F., Vineis, P., Tumino, R., Panico, S., Berglund, G., Simán, H., Nyren, O., Hallmans, G., Martinez, C., Dorronsoro, M., Barricarte, A., Navarro, C., Quiros, J. R., Allen, N., Key, T., Bingham, S., Caldas, C., Linseisen, J., Nagel, G., Overvad, K., Tjonneland, A., Boshuizen, H. C., Peeters, P. H., Numans, M. E., Clavel-Chapelon, F., Trichopoulou, A., Lund, E., Jenab, M., Kaaks, R., Riboli, E., Gonzalez, C. A. : DNA repair polymorphisms and the risk of stomach adenocarcinoma and severe chronic gastritis in the EPIC-EURGAST study. International Journal of Epidemiology, 37(6), 1316- 1325, 2008. https://doi.org/10.1093/ije/dyn145
  10. Kiyohara, C., Yoshimasu, K. : Genetic polymorphisms in the nucleotide excision repair pathway and lung cancer risk: a meta-analysis. International Journal of Medical Sciences, 4(2), 59-71, 2007.
  11. Han, J., Colditz, G. A., Liu, J. S., Hunter, D. J. : Genetic variation in XPD, sun exposure, and risk of skin cancer. Cancer Epidemiology Biomarkers & Prevention, 14(6), 1539-1544, 2005. https://doi.org/10.1158/1055-9965.EPI-04-0846
  12. Hung, R. J., Hall, J., Brennan, P., Boffetta, P. : Genetic polymorphisms in the base excision repair pathway and cancer risk: a HuGE review. American Journal of Epidemiology, 162(10), 925-942, 2005. https://doi.org/10.1093/aje/kwi318
  13. Li, H., Ha, T. C., Tai, B. C. : XRCC1 gene polymorphisms and breast cancer risk in different populations: a meta-analysis. The Breast, 18(3), 183-191, 2009. https://doi.org/10.1016/j.breast.2009.03.008
  14. Han, S., Zhang, H. T., Wang, Z., Xie, Y., Tang, R., Mao, Y., Li, Y. : DNA repair gene XRCC3 polymorphisms and cancer risk: a meta-analysis of 48 casecontrol studies. European Journal of Human Genetics, 14(10), 1136-1144, 2006. https://doi.org/10.1038/sj.ejhg.5201681
  15. Schulte, P. A., Petera, F P. : A conceptual and historical framework for molecular epidemiology. Molecular epidemiology: principles and practives, ed. Schulte, P. A. and Perera, F. P. San Dieago: Academic Press, 1993.
  16. Khoury, M. J., Beaty, T. H., Cohen, B. H. : Fundamentals of genetic epidemiology. New York: NY: Oxford University Press, 1993.
  17. Cuzick, J. : Interaction, subgroup analysis and sample size. Metabolic polymorphisms and susceptibility to cancer. ed. Vineis P., et al., Lyon: IARC Scientific Publishers, 1999.
  18. Carporaso, N. : Selection of candidate genes for population studies. in Metabolic polymorhisms and susceptibility to cancer, Vineis, P., et al., Editors, Lyon, 1999.
  19. Zheng, W. : Epidemiological studies of genetic factors for cancer. Fundamentals of cancer epidemiology, ed. Nasca PC and Pastides H. 2001: An Aspen Publications.
  20. Park, S. K., Lee, S. D. : Environmental pollutants and epigenetics. Korean Society of Environment Health, 35(5), 343-354, 2009. https://doi.org/10.5668/JEHS.2009.35.5.343
  21. IARC : IARC Monographs on the evaluation of carcinogenic risks to humans. 2009.
  22. National Toxicology Program, Carcinogens listed in the eleventh report, in 11th Report on Carcinogens, N.T. Program, Editor. 2005.
  23. Luch, A. : Nature and nurture - lessons from chemical carcinogenesis. Nature Reviews Cancer, 5(2), 113-125, 2005. https://doi.org/10.1038/nrc1546
  24. Wogan, G. N., Hecht, S. S., Felton, J. S., Conney, A. H., Loeb, L. A. : Environmental and chemical carcinogenesis. Seminars in Cancer Biology, 14(6), 473-486, 2004. https://doi.org/10.1016/j.semcancer.2004.06.010
  25. Poulsen, H. E., Loft, S., Wassermann, K. : Cancer risk related to genetic polymorphisms in carcinogen metabolism and DNA repair. Journal of Pharmacology and Toxicology, 72, 93-103, 1993. https://doi.org/10.1111/j.1600-0773.1993.tb01676.x
  26. Lash, L. H., Putt, D. A., Cai, H. : Drug metabolism enzyme expression and activity in primary cultures of human proximal tubular cells. Toxicology, 244(1), 56- 65, 2008. https://doi.org/10.1016/j.tox.2007.10.022
  27. Aliya, S ., R eddanna, P ., T hyagaraju, K . : Does glutathione S-transferase Pi (GST-Pi) a marker protein for cancer? Molecular and Cellular Biochemistry, 253(1), 319-327, 2003. https://doi.org/10.1023/A:1026036521852
  28. Taylor and Francis, in Glutathione S-transferases and carcinogenesis, Mantle, T. J., Pickett, C. B. and Hayes, J. D., Editors. New York, 1987.
  29. Talalay, P ., De Long, M. J ., P rochaska, H. J . : Identification of a common chemical signal regulating the induction of enzymes that protect against chemical carcinogenesis. Proceedings of the National Academy of Sciences USA, 85(21), 8261-8265, 1988. https://doi.org/10.1073/pnas.85.21.8261
  30. Modrich, P. : Mismatch repair, genetic stability and tumour avoidance. Philosophical Transactions of the Royal Society, 347(1319), 89-95, 1995. https://doi.org/10.1098/rstb.1995.0014
  31. Friedberg, E. C. : How nucleotide excision repair protects against cancer. Nature Reviews Cancer, 1(1), 22-33, 2001. https://doi.org/10.1038/35094000
  32. Bell, D. A., Taylor, J. A., Paulson, D. F., Robertson, C. N., Mohler, J. L., Lucier, G .W. : Genetic risk and carcinogen exposure: a common inherited defect of the carcinogen-metabolism gene glutathione S-transferase M1 (GSTM1) that increases susceptibility to bladder cancer. Journal of the National Cancer Institute, 85(14), 1159-1164, 1993. https://doi.org/10.1093/jnci/85.14.1159
  33. Engel, L. S., Taioli, E., Pfeiffer, R., Garcia-Closas, M., Marcus, P. M., Lan, Q., Boffetta, P., Vineis, P., Bell, D. A., Autrup, H., Branch, R. A., Brockmoller, J., Daly, A. K., Heckbert, S. R., Kalina, I., Kang, D., Katoh, T., Lafuente, A., Lin, H. J., Taylor, J. A., Romkes, M., Rothman, N. : Pooled analysis and meta-analysis of glutathione S-transferase M1 and bladder cancer: a HuGE review. American Journal of Epidemiology, 156(2), 95-109, 2002. https://doi.org/10.1093/aje/kwf018
  34. Han, J., Hankinson, S. E., Ranu, H., De Vivo, I., Hunter, D. J. : Polymorphisms in DNA double-strand break repair genes and breast cancer risk in the Nurses' Health Study. Carcinogenesis, 25(2), 189-195, 2004.
  35. Ratnasinghe, D., Yao, S. X., Tangrea, J. A., Qiao, Y. L., Andersen, M. R., Barrett, M. J., Giffen, C. A., Erozan, Y., Tockman, M. S., Taylor, P. R. : Polymorphisms of the DNA repair gene XRCC1 and lung cancer risk. Cancer Epidemiology Biomarkers & Prevention, 10(2), 119-123, 2001.
  36. Huang, W. Y., Olshan, A. F., Schwartz, S. M., Berndt, S. I., Chen, C., Llaca, V., Chanock, S. J., Fraumeni, J. F. Jr, Hayes, R. B. : Selected genetic polymorphisms in MGMT, XRCC1, XPD, and XRCC3 and risk of head and neck cancer: a pooled analysis. Cancer Epidemiology Biomarkers & Prevention, 14(7), 1747-1753, 2005. https://doi.org/10.1158/1055-9965.EPI-05-0162
  37. Guy, C. A., Hoogendoorn, B., Smith, S. K., Coleman, S., O'Donovan, M. C., Buckland, P. R. : Promoter polymorphisms in glutathione-S-transferase genes affect transcription. Pharmacogenetics, 14(1), 45-51, 2004. https://doi.org/10.1097/00008571-200401000-00005
  38. Johnson, A. D., Wang, D., Sadee, W. : Polymorphisms affecting gene regulation and mRNA processing: broad implications for pharmacogenetics. Pharmacology & Therapeutics, 106(1), 19-38, 2005. https://doi.org/10.1016/j.pharmthera.2004.11.001
  39. Lee, S. A. : Gene-diet interaction on cancer risk in epidemiological studies. Journal of Preventive Medicine and Public Health, 42(6), 360-370, 2009. https://doi.org/10.3961/jpmph.2009.42.6.360
  40. Taylor, R., Najafi, F., Dobson, A. : Meta-analysis of studies of passive smoking and lung cancer: effects of study type and continent. International Journal of Epidemiology, 36(5), 1048-1059, 2007. https://doi.org/10.1093/ije/dym158
  41. Howsam, M., Grimalt, J. O., Guino, E., Navarro, M., Marti-Rague, J., Peinado, M. A., Capella, G., Moreno, V. : Bellvitge Colorectal Cancer Group.: Organochlorine exposure and colorectal cancer risk. Environmental Health Perspectives, 112(15), 1460-1466, 2004. https://doi.org/10.1289/ehp.7143
  42. Infante-Rivard, C., Amre, D., Sinnett, D. : GSTT1 and CYP2E1 polymorphisms and trihalomethanes in drinking water: effect on childhood leukemia. Environmental Health Perspectives, 110, 591-593, 2003.
  43. Xiong, P., Bondy, M. L., Li, D., Shen, H., Wang, L. E., Singletary, S. E., Spitz, M. R., Wei, Q. : Sensitivity to benzo(a)pyrene diol-epoxide associated with risk of breast cancer in young women and modulation by glutathione S-transferase polymorphisms: a case-control study. Cancer Research, 61(23), 8465- 8469, 2001.
  44. Shinka, T., Ogura, H., Morita, T., Nishikawa, T., Fujinaga, T., Ohkawa, T. : Relationship between glutathione S-transferase M1 deficiency and urothelial cancer in dye workers exposed to aromatic amines. The Journal of Urology, 159(2), 380-383, 1998. https://doi.org/10.1016/S0022-5347(01)63924-7
  45. Ji, F., Wang, W., Xia, Z. L., Zheng, Y. J., Qiu, Y. L., Wu, F., Miao, W. B., Jin, R. F., Qian, J., Jin, L., Zhu, Y. L., Christiani, D. C. : Prevalence and persistence of chromosomal damage, and susceptible genotypes of metabolic and dna repair genes in chinese vinyl chloride-exposed workers. Carcinogenesis, [Epub ahead of print], 2010.
  46. Wang, S., Chanock, S., Tang, D., Li, Z., Jedrychowski, W., Perera, F. P. : Assessment of interactions between PAH exposure and genetic polymorphisms on PAHDNA adducts in African American, Dominican, and Caucasian mothers and newborns. Cancer Epidemiology Biomarkers & Prevention, 17(2), 405-413, 2008. https://doi.org/10.1158/1055-9965.EPI-07-0695
  47. da Silva, J., Moraes, C. R., Heuser, V. D., Andrade, V. M., Silva, F. R., Kvitko, K., Emmel, V., Rohr, P., Bordin, D. L., Andreazza, A. C., Salvador, M., Henriques, J. A., Erdtmann, B. : Evaluation of genetic damage in a Brazilian population occupationally exposed to pesticides and its correlation with polymorphisms in metabolizing genes. Mutagenesis, 23(5), 415-422, 2008. https://doi.org/10.1093/mutage/gen031
  48. Leng, S., Stidley, C. A., Bernauer, A. M., Picchi, M. A., Sheng, X., Frasco, M. A., Van Den Berg, D., Gilliland, F. D., Crowell, R. E., Belinsky, S. A. : Haplotypes of DNMT1 and DNMT3B are associated with mutagen sensitivity induced by benzo[a]pyrene diol epoxide among smokers. Carcinogenesis, 29(7), 1380-1385, 2008. https://doi.org/10.1093/carcin/bgn121
  49. Ketelslegers, H. B., Gottschalk, R. W., Koppen, G., Schoeters, G., Baeyens, W. F., van Larebeke, N. A., van Delft, J. H., Kleinjans, J. C. : Multiplex genotyping as a biomarker for susceptibility to carcinogenic exposure in the FLEHS biomonitoring study. Cancer Epidemiology Biomarkers & Prevention, 17(8), 1902- 1912, 2008. https://doi.org/10.1158/1055-9965.EPI-08-0045
  50. Pavanello, S., Pulliero, A., Siwinska, E., Mielzynska, D., Clonfero, E. : Reduced nucleotide excision repair and GSTM1-null genotypes influence anti-B[a]PDE DNA adduct levels in mononuclear white blood cells of highly PAH-exposed coke oven workers. Carcinogenesis, 26(1), 169-175, 2005.
  51. Lodovici, M., Luceri, C., Guglielmi, F., Bacci, C., Akpan, V., Fonnesu, M. L., Boddi, V., Dolara, P. : Benzo(a)pyrene diolepoxide (BPDE)-DNA adduct levels in leukocytes of smokers in relation to polymorphism of CYP1A1, GSTM1, GSTP1, GSTT1, and mEH. Cancer Epidemiology Biomarkers & Prevention, 13(8), 1342-1348, 2004.