全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
ISRN Oncology  2013 

Epigenetic Influences in the Aetiology of Cancers Arising from Breast and Prostate: A Hypothesised Transgenerational Evolution in Chromatin Accessibility

DOI: 10.1155/2013/624794

Full-Text   Cite this paper   Add to My Lib

Abstract:

Epidemiological studies have consistently supported the notion that environmental and/or dietary factors play a central role in the aetiology of cancers of the breast and prostate. However, for more than five decades investigators have failed to identify a single cause-and-effect factor, which could be implicated; identification of a causative entity would allow the implementation of an intervention strategy in at-risk populations. This suggests a more complex pathoaetiology for these cancer sites, compared to others. When one examines the increases or decreases in incidence of specific cancers amongst migrant populations, it is notable that disease arising in colon or stomach requires one or at most two generations to exhibit a change in incidence to match that of high-incidence regions, whereas for breast or prostate cancer, at least three generations are required. This generational threshold could suggest a requirement for nonmutation-driven epigenetic alterations in the F0/F1 generations (parental/offspring adopting a more westernized lifestyle), which then predisposes the inherited genome of subsequent generations to mutagenic/genotoxic alterations leading to the development of sporadic cancer in these target sites. As such, individual susceptibility to carcinogen insult would not be based per se on polymorphisms in activating/detoxifying/repair enzymes, but on elevated accessibility of crucial target genes (e.g., oncogenes, tumour suppressor genes) or hotspots therein to mutation events. This could be termed a genomic susceptibility organizational structure (SOS). Several exposures including alcohol and heavy metals are epigens (i.e., modifiers of the epigenome), whereas others are mutagenic/genotoxic, for example, heterocyclic aromatic amines; humans are continuously and variously exposed to mixtures of these agents. Within such a transgenerational multistage model of cancer development, determining the interaction between epigenetic modification to generate a genomic SOS and genotoxic insult will facilitate a new level of understanding in the aetiology of cancer. 1. Introduction Epidemiological studies clearly implicate environmental and/or lifestyle factors in the aetiology of cancers arising in hormone-responsive tissues, such as those from the breast or prostate [1]. This is based on the observations that incidence of these cancers is high in regions such as Northern/Western Europe and the USA, whereas recorded levels in other areas including China and India are traditionally some 10-fold lower [2] (Figure 1(a)). However, when populations

References

[1]  P. L. Grover and F. L. Martin, “The initiation of breast and prostate cancer,” Carcinogenesis, vol. 23, no. 7, pp. 1095–1102, 2002.
[2]  D. M. Parkin, P. Pisani, and J. Ferlay, “Estimates of the worldwide incidence of 25 major cancers in 1990,” International Journal of Cancer, vol. 80, no. 6, pp. 827–841.
[3]  C. S. Muir, J. Nectoux, and J. Staszewski, “The epidemiology of prostatic cancer. Geographical distribution and time-trends,” Acta Oncologica, vol. 30, no. 2, pp. 133–140, 1991.
[4]  J. Peto, “Cancer epidemiology in the last century and the next decade,” Nature, vol. 411, no. 6835, pp. 390–395, 2001.
[5]  M. Saito, M. Matsuzaki, T. Sakuma, et al., “Clinicopathological study of non-palpable familial breast cancer detected by screening mammography and diagnosed as DCIS,” Breast Cancer. In press.
[6]  F. Stenback, R. Peto, and P. Shubik, “Initiation and promotion at different ages and doses in 2200 mice. III. Linear extrapolation from high doses may underestimate low-dose tumour risks,” British Journal of Cancer, vol. 44, no. 1, pp. 24–34, 1981.
[7]  A. L. Reddy and P. J. Fialkow, “Influence of dose of initiator on two-stage skin carcinogenesis in BALB/c mice with cellular moscaicism,” Carcinogenesis, vol. 9, no. 5, pp. 751–754, 1988.
[8]  A. L. Herbst, R. E. Scully, and S. J. Robboy, “The significance of adenosis and clear cell adenocarcinoma of the genital tract in young females,” Journal of Reproductive Medicine for the Obstetrician and Gynecologist, vol. 15, no. 1, pp. 5–11, 1975.
[9]  D. C. Dolinoy, J. R. Weidman, R. A. Waterland, and R. L. Jirtle, “Maternal genistein alters coat color and protects Avy mouse offspring from obesity by modifying the fetal epigenome,” Environmental Health Perspectives, vol. 114, no. 4, pp. 567–572, 2006.
[10]  M. Ur Rehman, Q. M. Buttar, M. Irfan-ul-Haq Khawaja, and M. Rizwan-ul-Haq Khawaja, “An impending cancer crisis in developing countries: are we ready for the challenge?” Asian Pacific Journal of Cancer Prevention, vol. 10, no. 4, pp. 719–720, 2009.
[11]  K. K. Carroll, E. B. Gammal, and E. R. Plunkett, “Dietary fat and mammary cancer,” Canadian Medical Association journal, vol. 98, no. 12, pp. 590–594, 1968.
[12]  F. L. Martin, P. L. Carmichael, C. Crofton-Sleigh, S. Venitt, D. H. Phillips, and P. L. Grover, “Genotoxicity of human mammary lipid,” Cancer Research, vol. 56, no. 23, pp. 5342–5346, 1996.
[13]  F. L. Martin, S. Venitt, P. L. Carmichael et al., “DNA damage in breast epithelial cells: detection by the single-cell gel (comet) assay and induction by human mammary lipid extracts,” Carcinogenesis, vol. 18, no. 12, pp. 2299–2305, 1997.
[14]  Y. L. Bronner, “Nutritional status outcomes for children: ethnic, cultural, and environmental contexts,” Journal of the American Dietetic Association, vol. 96, no. 9, pp. 891–903, 1996.
[15]  C. Maringe, P. Mangtani, B. Rachet, D. A. Leon, M. P. Coleman, and I. dos Santos Silva, “Cancer incidence in South Asian migrants to England, 1986–2004: unraveling ethnic from socioeconomic differentials,” International Journal of Cancer. In press.
[16]  F. L. Martin, “Epigenomics and disease, 10th anniversary winter meeting of the UK Molecular Epidemiology Group (MEG), the Royal Statistical Society, London, UK, 8th December 2006,” Mutagenesis, vol. 22, no. 6, pp. 425–427, 2007.
[17]  V. V. Lao and W. M. Grady, “Epigenetics and colorectal cancer,” Nature Reviews Gastroenterology and Hepatology, vol. 8, no. 12, pp. 686–700, 2011.
[18]  L. Hou, X. Zhang, D. Wang, and A. Baccarelli, “Environmental chemical exposures and human epigenetics,” International Journal of Epidemiology, vol. 41, no. 1, pp. 79–105, 2012.
[19]  A. Merlo, J. G. Herman, L. Mao et al., “ CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers,” Nature Medicine, vol. 1, no. 7, pp. 686–692, 1995.
[20]  A. L. Reed, J. Califano, P. Cairns et al., “High frequency of p16 (CDKN2/MTS-1/INK4A) inactivation in head and neck squamous cell carcinoma,” Cancer Research, vol. 56, no. 16, pp. 3630–3633, 1996.
[21]  J. G. Herman, C. I. Civin, J. P. J. Issa, M. I. Collector, S. J. Sharkis, and S. B. Baylin, “Distinct patterns of inactivation of p15INK4B and p16INK4A characterize the major types of hematological malignancies,” Cancer Research, vol. 57, no. 5, pp. 837–841, 1997.
[22]  S. Snellenberg, L. M. Strooper, A. T. Hesselink, et al., “Development of a multiplex methylation-specific PCR as candidate triage test for women with an HPV-positive cervical scrape,” BMC Cancer, vol. 12, no. 1, article 551, 2012.
[23]  J. W. Kim, S. T. Kim, A. R. Turner, et al., “Identification of new differentially methylated genes that have potential functional consequences in prostate cancer,” PloS One, vol. 7, no. 10, Article ID e48455, 2012.
[24]  H. Heyn, F. J. Carmona, A. Gomez, et al., “DNA methylation profiling in breast cancer discordant identical twins identifies DOK7 as novel epigenetic biomarker,” Carcinogenesis, vol. 34, no. 1, pp. 102–108, 2013.
[25]  B. Schuster-B?ckler and B. Lehner, “Chromatin organization is a major influence on regional mutation rates in human cancer cells,” Nature, vol. 488, no. 7411, pp. 504–507, 2012.
[26]  O. A. Botrugno, T. Robert, F. Vanoli, M. Foiani, and S. Minucci, “Molecular pathways: old drugs define new pathways: non-histone acetylation at the crossroads of the DNA damage response and autophagy,” Clinical Cancer Research, vol. 18, no. 9, pp. 2436–2442, 2012.
[27]  M. Zeybel, T. Hardy, Y. K. Wong, et al., “Multigenerational epigenetic adaptation of the hepatic wound-healing response,” Nature Medicine, vol. 18, no. 9, pp. 1369–1377, 2012.
[28]  C. Lu and C. B. Thompson, “Metabolic regulation of epigenetics,” Cell Metabolism, vol. 16, no. 1, pp. 9–17, 2012.
[29]  E. B. Keverne, “Epigenetics and brain evolution,” Epigenomics, vol. 3, no. 2, pp. 183–191, 2011.
[30]  F. Sanchis-Gomar, J. L. Garcia-Gimenez, C. Perez-Quilis, M. C. Gomez-Cabrera, F. V. Pallardo, and G. Lippi, “Physical exercise as an epigenetic modulator: eustress, the “positive stress” as an effector of gene expression,” The Journal of Strength & Conditioning Research, vol. 26, no. 12, pp. 3469–3472, 2012.
[31]  R. Maruyama and H. Suzuki, “Long noncoding RNA involvement in cancer,” BMB Reports, vol. 45, no. 11, pp. 604–611, 2012.
[32]  A. Banerjee and K. Luettich, “MicroRNAs as potential biomarkers of smoking-related diseases,” Biomarkers in Medicine, vol. 6, no. 5, pp. 671–684, 2012.
[33]  S. Volinia, G. A. Calin, C. G. Liu et al., “A microRNA expression signature of human solid tumors defines cancer gene targets,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 7, pp. 2257–2261, 2006.
[34]  O. Meikar, M. Da Ros, and N. Kotaja, “Epigenetic regulation of male germ cell differentiation,” Subcellular Biochemistry, vol. 61, pp. 119–138, 2012.
[35]  S. Malan-Müller, S. M. Hemmings, and S. Seedat, “Big effects of small RNAs: a review of microRNAs in anxiety,” Molecular Neurobiology. In press.
[36]  M. Ballarino, L. Jobert, D. Dembélé, P. de la Grange, D. Auboeuf, and L. Tora, “TAF15 is important for cellular proliferation and regulates the expression of a subset of cell cycle genes through miRNAs,” Oncogene. In press.
[37]  M. A. Listowski, E. Heger, D. M. Bogus?awska, et al., “microRNAs: fine tuning of erythropoiesis,” Cellular and Molecular Biology Letters, vol. 18, no. 1, pp. 34–46, 2013.
[38]  A. Brevik, B. Lindeman, G. Brunborg, and N. Duale, “Paternal benzo[a]pyrene exposure modulates microRNA expression patterns in the developing mouse embryo,” International Journal of Cell Biology, vol. 2012, Article ID 407431, 11 pages, 2012.
[39]  K. A. Lillycrop and G. C. Burdge, “Epigenetic mechanisms linking early nutrition to long term health,” Best Practice & Research Clinical Endocrinology & Metabolism, vol. 26, no. 5, pp. 667–676, 2012.
[40]  K. A. Lillycrop, “Effect of maternal diet on the epigenome: implications for human metabolic disease,” Proceedings of the Nutrition Society, vol. 70, no. 1, pp. 64–72, 2011.
[41]  M. Lechner, C. Boshoff, and S. Beck, “Cancer Epigenome,” Advances in Genetics, vol. 70, pp. 247–276, 2010.
[42]  J. A. Williams, F. L. Martin, G. H. Muir, A. Hewer, P. L. Grover, and D. H. Phillips, “Metabolic activation of carcinogens and expression of various cytochromes P450 in human prostate tissue,” Carcinogenesis, vol. 21, no. 9, pp. 1683–1689, 2000.
[43]  F. L. Martin, I. I. Patel, O. Sozeri et al., “Constitutive expression of bioactivating enzymes in normal human prostate suggests a capability to activate pro-carcinogens to DNA-damaging metabolites,” Prostate, vol. 70, no. 14, pp. 1586–1599, 2010.
[44]  N. Ragavan, R. Hewitt, L. J. Cooper et al., “CYP1B1 expression in prostate is higher in the peripheral than in the transition zone,” Cancer Letters, vol. 215, no. 1, pp. 69–78, 2004.
[45]  K. Abass, V. L?msa, P. Reponen, et al., “Characterization of human cytochrome P450 induction by pesticides,” Toxicology, vol. 294, no. 1, pp. 17–26, 2012.
[46]  M. F. Fraga, E. Ballestar, M. F. Paz et al., “Epigenetic differences arise during the lifetime of monozygotic twins,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 30, pp. 10604–10609, 2005.
[47]  K. Szarc vel Szic, M. N. Ndlovu, G. Haegeman, and W. Vanden Berghe, “Nature or nurture: let food be your epigenetic medicine in chronic inflammatory disorders,” Biochemical Pharmacology, vol. 80, no. 12, pp. 1816–1832, 2010.
[48]  M. D. Anway, A. S. Cupp, N. Uzumcu, and M. K. Skinner, “Toxicology: epigenetic transgenerational actions of endocrine disruptors and male fertility,” Science, vol. 308, no. 5727, pp. 1466–1469, 2005.
[49]  H. Bartsch and R. Montesano, “Relevance of nitrosamines to human cancer,” Carcinogenesis, vol. 5, no. 11, pp. 1381–1393, 1984.
[50]  D. A. Gouas, S. Villar, S. Ortiz-Cuaran, et al., “TP53 R249S mutation, genetic variations in HBX and risk of hepatocellular carcinoma in The Gambia,” Carcinogenesis, vol. 33, no. 6, pp. 1219–1224, 2012.
[51]  M. G?ttlicher, S. Minucci, P. Zhu et al., “Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells,” The EMBO Journal, vol. 20, no. 24, pp. 6969–6978, 2002.
[52]  T. Ahmad, K. Shekh, S. Khan, et al., “Pretreatment of valproic acid, a histone deacetylase inhibitor enhances the sensitivity of peripheral blood micronucleus assay in rodents,” Mutation Research. In press.
[53]  A. T. Vo and R. M. Millis, “Epigenetics and breast cancers,” Obstetrics and Gynecology International, vol. 2012, Article ID 602720, 10 pages, 2012.
[54]  R. Martinez-Zamudio and H. C. Ha, “Environmental epigenetics in metal exposure,” Epigenetics, vol. 6, no. 7, pp. 820–827, 2011.
[55]  Y. Arai, J. Ohgane, S. Yagi, et al., “Epigenetic assessment of environmental chemicals detected in maternal peripheral and cord blood samples,” Journal of Reproduction and Development, vol. 57, no. 4, pp. 507–517, 2011.
[56]  F. L. Martin, “Complex mixtures that may contain mutagenic and/or genotoxic components: a need to assess in vivo target-site effect(s) associated with in vitro-positive(s),” Chemosphere, vol. 69, no. 6, pp. 841–848, 2007.
[57]  V. Llabjani, J. Trevisan, K. C. Jones, R. F. Shore, and F. L. Martin, “Binary mixture effects by PBDE congeners (47, 153, 183, or 209) and PCB congeners (126 or 153) in MCF-7 cells: biochemical alterations assessed by IR spectroscopy and multivariate analysis,” Environmental Science and Technology, vol. 44, no. 10, pp. 3992–3998, 2010.
[58]  L. J. Lister, C. Svendsen, J. Wright, H. L. Hooper, and D. J. Spurgeon, “Modelling the joint effects of a metal and a pesticide on reproduction and toxicokinetics in Lumbricid earthworms,” Environment International, vol. 37, no. 4, pp. 663–670, 2011.
[59]  J. A. Rusiecki, A. Baccarelli, V. Bollati, L. Tarantini, L. E. Moore, and E. C. Bonefeld-Jorgensen, “Global DNA hypomethylation is associated with high serum-persistent organic pollutants in Greenlandic inuit,” Environmental Health Perspectives, vol. 116, no. 11, pp. 1547–1552, 2008.
[60]  Y. Yuasa, “Epigenetics in molecular epidemiology of cancer: a new scope,” Advances in Genetics, vol. 71, pp. 212–235, 2010.
[61]  A. Janesick and B. Blumberg, “Obesogens, stem cells and the developmental programming of obesity,” International Journal of Andrology, vol. 35, no. 3, pp. 437–448, 2012.
[62]  E. Karoutsou and A. Polymeris, “Environmental endocrine disruptors and obesity,” Endocrine Regulations, vol. 46, no. 1, pp. 37–46, 2012.
[63]  B. C. Christensen and C. J. Marsit, “Epigenomics in environmental health,” Frontiers in Genetics, vol. 2, article 84, 2011.
[64]  T. Wang, J. G. Garcia, and W. Zhang, “Epigenetic regulation in particulate matter-mediated cardiopulmonary toxicities: a systems biology perspective,” Current Pharmacogenomics and Personalized Medicine, vol. 10, no. 4, pp. 314–321, 2012.
[65]  M. Tian, S. Peng, F. L. Martin, et al., “Perfluorooctanoic acid induces gene promoter hypermethylation of glutathione-S-transferase Pi in human liver L02 cells,” Toxicology, vol. 296, pp. 48–55, 2012.
[66]  R. Feil and M. F. Fraga, “Epigenetics and the environment: emerging patterns and implications,” Nature Reviews Genetics, vol. 13, no. 2, pp. 97–109, 2012.
[67]  J. R. Roberts, C. J. Karr, and Council on Environmental Health, “Pesticide exposure in children,” Pediatrics, vol. 130, no. 6, pp. e1765–e1788, 2012.
[68]  I. P. Pogribny and I. Rusyn, “Environmental toxicants, epigenetics, and cancer,” Advances in Experimental Medicine and Biology, vol. 754, pp. 215–232, 2013.
[69]  B. Wang, Y. Li, C. Shao, Y. Tan, and L. Cai, “Cadmium and its epigenetic effects,” Current Medicinal Chemistry, vol. 19, no. 16, pp. 2611–2620, 2012.
[70]  S. Singh and S. S. . Li, “Epigenetic effects of environmental chemicals bisphenol A and phthalates,” International Journal of Molecular Sciences, vol. 13, no. 8, pp. 10143–10153, 2012.
[71]  C. M. Markey, P. R. Wadia, B. S. Rubin, C. Sonnenschein, and A. M. Soto, “Long-term effects of fetal exposure to low doses of the xenoestrogen bisphenol-A in the female mouse genital tract,” Biology of Reproduction, vol. 72, no. 6, pp. 1344–1351, 2005.
[72]  A. F. Fleisch, R. O. Wright, and A. A. Baccarelli, “Environmental epigenetics: a role in endocrine disease,” Journal of Molecular Endocrinology, vol. 49, no. 2, pp. R61–R67, 2012.
[73]  R. K. Vempati, “DNA damage in the presence of chemical genotoxic agents induce acetylation of H3K56 and H4K16 but not H3K9 in mammalian cells,” Molecular Biology Reports, vol. 39, no. 1, pp. 303–308, 2012.
[74]  M. Esteller, “Relevance of DNA methylation in the management of cancer,” The Lancet Oncology, vol. 4, no. 6, pp. 351–358, 2003.
[75]  M. Kim, M. Bae, H. Na, and M. Yang, “Environmental toxicants-induced epigenetic alterations and their reversers,” Journal of Environmental Science and Health, Part C, vol. 30, no. 4, pp. 323–367, 2012.
[76]  M. Talikka, N. Sierro, N. V. Ivanov, et al., “Genomic impact of cigarette smoke, with application to three smoking-related diseases,” Critical Reviews in Toxicology, vol. 42, no. 10, pp. 877–889, 2012.
[77]  C. J. Mattingly, T. E. McKone, M. A. Callahan, J. A. Blake, and E. A. Hubal, “Providing the missing link: the exposure science ontology ExO,” Environmental Science & Technology, vol. 46, no. 6, pp. 3046–3053, 2012.
[78]  C. J. Steves, T. D. Spector, and S. H. Jackson, “Ageing, genes, environment and epigenetics: what twin studies tell us now, and in the future,” Age and Ageing, vol. 41, no. 5, pp. 581–586, 2012.
[79]  H. Heyn, N. Li, H. J. Ferreira, et al., “Distinct DNA methylomes of newborns and centenarians,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 26, pp. 10522–10527, 2012.
[80]  H. H. Nelson, C. J. Marsit, B. C. Christensen, et al., “Key epigenetic changes associated with lung cancer development: results from dense methylation array profiling,” Epigenetics, vol. 7, no. 6, pp. 559–566, 2012.
[81]  P. C. Turner, A. C. Collinson, Y. B. Cheung et al., “Aflatoxin exposure in utero causes growth faltering in Gambian infants,” International Journal of Epidemiology, vol. 36, no. 5, pp. 1119–1125, 2007.
[82]  M. Verma, “Epigenetic biomarkers in cancer epidemiology,” Methods in Molecular Biology, vol. 863, pp. 467–480, 2012.
[83]  P. Vineis and D. Kriebel, “Causal models in epidemiology: past inheritance and genetic future,” Environmental Health, vol. 5, article 21, 2006.
[84]  D. Simmons, D. R. R. Williams, and M. J. Powell, “The Coventry Diabetes Study: prevalence of diabetes and impaired glucose tolerance in Europids and Asians,” Quarterly Journal of Medicine, vol. 81, no. 296, pp. 1021–1030, 1991.
[85]  J. Dhawan, C. L. Bray, R. Warburton, D. S. Ghambhir, and J. Morris, “Insulin resistance, high prevalence of diabetes, and cardiovascular risk in immigrant Asians,” British Heart Journal, vol. 72, no. 5, pp. 413–421, 1994.
[86]  M. C. Carey and B. Paigen, “Epidemiology of the American Indians' burden and its likely genetic origins,” Hepatology, vol. 36, no. 4, part 1, pp. 781–791, 2002.
[87]  H. C. Pitot, “The stability of events in the natural history of neoplasia,” American Journal of Pathology, vol. 89, no. 3, pp. 703–716, 1977.
[88]  I. Chouroulinkov, A. Gentil, and B. Tierney, “Biological activities of dihydrodiols derived from two polycyclic hydrocarbons in rodent test systems,” British Journal of Cancer, vol. 39, no. 4, pp. 376–382, 1979.
[89]  R. A. CASE, “Incidence of death from tumours of the urinary bladder,” British Journal of Preventive & Social Medicine, vol. 7, no. 1, pp. 14–19, 1953.
[90]  R. A. CASE, “The expected frequency of bladder tumour in works populations,” British Journal of iIndustrial Medicine, vol. 10, no. 2, pp. 114–120, 1953.
[91]  R. Doll and R. Peto, “Mortality in relation to smoking: 20 years' observations on male British doctors,” British Medical Journal, vol. 2, no. 6051, pp. 1525–1536, 1976.
[92]  R. Doll and R. Peto, “Cigarette smoking and bronchial carcinoma: dose and time relationships among regular smokers and lifelong non-smokers,” Journal of Epidemiology and Community Health, vol. 32, no. 4, pp. 303–313, 1978.
[93]  I. C. Hsu, R. A. Metcalf, T. Sun, J. A. Welsh, N. J. Wang, and C. C. Harris, “Mutational hotspot in the p53 gene in human hepatocellular carcinomas,” Nature, vol. 350, no. 6317, pp. 427–428, 1991.
[94]  M. C. Hollstein, C. P. Wild, F. Bleicher et al., “p53 Mutations and aflatoxin B1 exposure in hepatocellular carcinoma patients from Thailand,” International Journal of Cancer, vol. 53, no. 1, pp. 51–55, 1993.
[95]  D. H. Phillips, “Fifty years of benzo(a)pyrene,” Nature, vol. 303, no. 5917, pp. 468–472, 1983.
[96]  W. Pfau, F. L. Martin, K. J. Cole et al., “Heterocyclic aromatic amines induce DNA strand breaks and cell transformation,” Carcinogenesis, vol. 20, no. 4, pp. 545–551, 1999.
[97]  M. Yamada, K. Kodama, S. Fujita et al., “Prevalence of skin neoplasms among the atomic bomb survivors,” Radiation Research, vol. 146, no. 2, pp. 223–226, 1996.
[98]  R. Doll, “Mortality from lung cancer in asbestos workers,” British Journal of Industrial Medicine, vol. 12, no. 2, pp. 81–86, 1955.
[99]  L. Rushton, S. J. Hutchings, L. Fortunato, et al., “Occupational cancer burden in Great Britain,” British Journal of Cancer, vol. 107, supplement 1, pp. 3–7, 2012.
[100]  D. H. Philips, “Understanding the genotoxicity of tamoxifen?” Carcinogenesis, vol. 22, no. 6, pp. 839–849, 2001.
[101]  J. Bendaly, K. J. Metry, M. A. Doll et al., “Role of human CYP1A1 and NAT2 in 2-amino-1-methyl-6-phenylimidazo[4,5-b] pyridine-induced mutagenicity and DNA adducts,” Xenobiotica, vol. 39, no. 5, pp. 399–406, 2009.
[102]  G. Maenhaut-Michel, R. Janel-Bintz, N. Samuel, and R. P. P. Fuchs, “Adducts formed by the food mutagen 2-amino-3-methylimidazo (4, 5-f) quinoline induce frameshift mutations at hot spots through an SOS-independent pathway,” Molecular and General Genetics, vol. 253, no. 5, pp. 634–641, 1997.
[103]  J. Bauer, G. Xing, H. Yagi, J. M. Sayer, D. M. Jerina, and H. Ling, “A structural gap in Dpo4 supports mutagenic bypass of a major benzo [a]pyrene dG adduct in DNA through template misalignment,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 38, pp. 14905–14910, 2007.
[104]  S. Saeed, C. Logie, K. J. Francoijs, et al., “Chromatin accessibility, p300, and histone acetylation define PML-RARa and AML1-ETO binding sites in acute myeloid leukemia,” Blood, vol. 120, no. 15, pp. 3058–3068, 2012.
[105]  N. Nalabothula and F. Carrier, “Cancer cells' epigenetic composition and predisposition to histone deacetylase inhibitor sensitization,” Epigenomics, vol. 3, no. 2, pp. 145–155, 2011.
[106]  A. Merrifield and W. Smith, “Sample size calculations for the design of health studies: a review of key concepts for non-statisticians,” New South Wales Public Health Bulletin, vol. 23, pp. 142–147, 2012.
[107]  B. Armstrong, “A simple estimator of minimum detectable relative risk, sample size, or power in cohort studies,” American Journal of Epidemiology, vol. 126, no. 2, pp. 356–358, 1987.
[108]  L. Ehrenberg and M. T?rnqvist, “Use of biomarkers in epidemiology: quantitative aspects,” Toxicology Letters, vol. 64, pp. 485–492, 1992.
[109]  P. Vineis, “The use of biomarkers in epidemiology: the example of bladder cancer,” Toxicology Letters, vol. 64-65, pp. 463–467, 1992.
[110]  F. P. Perera, L. A. Mooney, M. Stampfer et al., “Associations between carcinogen-DNA damage, glutathione S-transferase genotypes, and risk of lung cancer in the prospective Physicians' Health Cohort Study,” Carcinogenesis, vol. 23, no. 10, pp. 1641–1646, 2002.
[111]  W. J. Fu, A. J. Stromberg, K. Viele, R. J. Carroll, and G. Wu, “Statistics and bioinformatics in nutritional sciences: analysis of complex data in the era of systems biology,” Journal of Nutritional Biochemistry, vol. 21, no. 7, pp. 561–572, 2010.
[112]  Z. Wu and H. Zhao, “Statistical power of model selection strategies for genome-wide association studies,” PLoS Genetics, vol. 5, no. 7, Article ID e1000582, 2009.
[113]  K. John, N. Ragavan, M. M. Pratt et al., “Quantification of phase I/II metabolizing enzyme gene expression and polycyclic aromatic hydrocarbon-DNA adduct levels in human prostate,” Prostate, vol. 69, no. 5, pp. 505–519, 2009.
[114]  P. R. Burton, A. L. Hansell, I. Fortier et al., “Size matters: just how big is BIG?: quantifying realistic sample size requirements for human genome epidemiology,” International Journal of Epidemiology, vol. 38, no. 1, pp. 263–273, 2009.
[115]  R. Doll, “Nature and nurture: possibilities for cancer control,” Carcinogenesis, vol. 17, no. 2, pp. 177–184, 1996.
[116]  T. Carreón, A. M. Ruder, P. A. Schulte, et al., “NAT2 slow acetylation and bladder cancer in workers exposed to benzidine,” International Journal of Cancer, vol. 118, no. 1, pp. 161–168, 2006.
[117]  M. C. Miller, H. W. Mohrenweiser, and D. A. Bell, “Genetic variability in susceptibility and response to toxicants,” Toxicology Letters, vol. 120, no. 1–3, pp. 269–280, 2001.
[118]  M. Ingelman-Sundberg, “Polymorphism of cytochrome P450 and xenobiotic toxicity,” Toxicology, vol. 181-182, pp. 447–452, 2002.
[119]  L. R. Kidd, D. W. Hein, K. Woodson et al., “Lack of association of the N-acetyltransferase NAT1*10 allele with prostate cancer incidence, grade, or stage among smokers in Finland,” Biochemical Genetics, vol. 49, no. 1-2, pp. 73–82, 2011.
[120]  Y. Lin, K. Yagyu, N. Egawa et al., “An overview of genetic polymorphisms and pancreatic cancer risk in molecular epidemiologic studies,” Journal of Epidemiology, vol. 21, no. 1, pp. 2–12, 2011.
[121]  B. K. Duncan and J. H. Miller, “Mutagenic deamination of cytosine residues in DNA,” Nature, vol. 287, no. 5782, pp. 560–561, 1980.
[122]  C. Schmutte, A. S. Yang, T. T. Nguyen, R. W. Beart, and P. A. Jones, “Mechanisms for the involvement of DNA methylation in colon carcinogenesis,” Cancer Research, vol. 56, no. 10, pp. 2375–2381, 1996.
[123]  S. E. Steck, M. M. Gaudet, J. A. Britton et al., “Interactions among GSTM1, GSTT1 and GSTP1 polymorphisms, cruciferous vegetable intake and breast cancer risk,” Carcinogenesis, vol. 28, no. 9, pp. 1954–1959, 2007.
[124]  C. A. Gonzalez, E. Riboli, K. Overvad et al., “Diet and cancer prevention: contributions from the European Prospective Investigation into Cancer and Nutrition (EPIC) study,” European Journal of Cancer, vol. 46, no. 14, pp. 2555–2562, 2010.
[125]  G. Masala, M. Assedi, B. Bendinelli, et al., “Fruit and vegetables consumption and breast cancer risk: the EPIC Italy study,” Breast Cancer Research and Treatment, vol. 132, no. 3, pp. 1127–1136, 2012.
[126]  D. Palli, G. Masala, P. Vineis et al., “Biomarkers of dietary intake of micronutrients modulate DNA adduct levels in healthy adults,” Carcinogenesis, vol. 24, no. 4, pp. 739–746, 2003.
[127]  M. Esteller, “The necessity of a human epigenome project,” Carcinogenesis, vol. 27, no. 6, pp. 1121–1125, 2006.
[128]  M. Kulis and M. Esteller, “DNA methylation and cancer,” Advances in Genetics, vol. 70, pp. 27–56, 2010.
[129]  S. Beck, “Taking the measure of the methylome,” Nature Biotechnology, vol. 28, no. 10, pp. 1026–1028, 2010.
[130]  A. Murrell, V. K. Rakyan, and S. Beck, “From genome to epigenome,” Human Molecular Genetics, vol. 14, no. 1, pp. R3–R10, 2005.
[131]  L. Nonn, V. Ananthanarayanan, and P. H. Gann, “Evidence for field cancerization of the prostate,” Prostate, vol. 69, no. 13, pp. 1470–1479, 2009.
[132]  D. J. Vander Griend, J. D'Antonio, B. Gurel, L. Antony, A. M. DeMarzo, and J. T. Isaacs, “Cell-autonomous intracellular androgen receptor signaling drives the growth of human prostate cancer initiating cells,” Prostate, vol. 70, no. 1, pp. 90–99, 2010.
[133]  C. Tetta, E. Ghigo, L. Silengo, M. C. Deregibus, and G. Camussi, “Extracellular vesicles as an emerging mechanism of cell-to-cell communication,” Endocrine. In press.
[134]  Y. Lee, S. El Andaloussi, and M. J. Wood, “Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy,” Human Molecular Genetics, vol. 21, no. 1, pp. R125–R134, 2012.
[135]  E. Gyorffy, L. Anna, K. Kovács, P. Rudnai, and B. Schoket, “Correlation between biomarkers of human exposure to genotoxins with focus on carcinogen-DNA adducts,” Mutagenesis, vol. 23, no. 1, pp. 1–18, 2008.
[136]  Y. M. D. Lo, “Fetal DNA in maternal plasma: progress through epigenetics,” Annals of the New York Academy of Sciences, vol. 1075, pp. 74–80, 2006.
[137]  J. G. Kelly, G. M. Najand, and F. L. Martin, “Characterisation of DNA methylation status using spectroscopy (mid-IR versus Raman) with multivariate analysis,” Journal of Biophotonics, vol. 4, no. 5, pp. 345–354, 2011.
[138]  B. N. Ames and L. S. Gold, “Paracelsus to parascience: the environmental cancer distraction,” Mutation Research, vol. 447, no. 1, pp. 3–13, 2000.
[139]  S. P. Hussain, L. J. Hofseth, and C. C. Harris, “Radical causes of cancer,” Nature Reviews Cancer, vol. 3, no. 4, pp. 276–285, 2003.

Full-Text

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133