We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Genetic variations associated with interindividual sensitivity in the response to arsenic exposure

    Alba Hernández

    Departament de Genètica i de Microbiologia, Edifici Cn, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.

    CIBER Epidemiologia y Salud Publica (CIBERESP), Spain

    &
    Ricard Marcos

    † Author for correspondence

    Departament de Genètica i de Microbiologia, Edifici Cn, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.

    CIBER Epidemiologia y Salud Publica (CIBERESP), Spain

    People are exposed to arsenic compounds environmentally, occupationally or therapeutically. In some areas, where arsenic is present in high proportions in the drinking water, this exposure represents an important health concern. Chronic exposure to arsenic leads to hyperkeratosis and loss of skin pigmentation, as well as to significant increases of different types of cancer in skin, lung, bladder and liver; in addition, other pathologies, such as vascular diseases, hepatotoxicity and diabetes, have also been related to arsenic exposure. Since high interindividual variability is observed among people exposed to equivalent doses, genetic susceptibility factors have been postulated to be involved. When inorganic arsenic enters into the body it undergoes metabolic conversion, in a process where methylation plays a crucial role. Trivalent forms, both inorganic and organic, are the most toxic and genotoxic and, for this reason, metabolic variations owing to variant alleles in genes involved in such a process have been the aim of several studies. Genes involved in other mechanisms, such as antioxidant defense and DNA-repair lesions, among others, have also been the subject of association studies. A survey of those studies related to individual susceptibility is summarized here. Results with genes involved in folate one-carbon metabolism and in arsenic transport across the cell membrane provide promising data for future studies.

    Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.

    Bibliography

    • Abernathy CO, Liu YP, Longfellow D et al.: Arsenic: health effects, mechanisms of actions, and research issues. Environ. Health Perspect.107,593–597 (1999).
    • Astolfi E, Maccagno A, Fernandez JC, Vaccara R, Stimola R: Review: relation between arsenic in drinking water and skin cancer. Biol. Trace Elem. Res.3,133–143 (1981).
    • Taylor PR, Qiao YL, Schatzkin A et al.: Relation of arsenic exposure to lung cancer among tin miners in Yunnan Province, China. Br. J. Ind. Med.46,881–886 (1989).
    • Chen WT, Hung WC, Kang WY, Huang YC, Chai CY: Urothelial carcinomas arising in arsenic-contaminated areas are associated with hypermethylation of the gene promoter of the death-associated protein kinase. Histopathology51,785–792 (2007).
    • Steinmaus C, Yuan Y, Bates MN, Smith AH: Case–control study of bladder cancer and drinking water arsenic in the western United States. Am. J. Epidemiol.158,1193–1201 (2003).
    • Bates MN, Rey OA, Biggs ML et al.: Case–control study of bladder cancer and exposure to arsenic in Argentina. Am. J. Epidemiol.159,381–389 (2004).
    • Chen CJ, Wang SL, Chiou JM et al.: Arsenic and diabetes and hypertension in human populations: a review. Toxicol. Appl. Pharmacol.222,298–304 (2007).
    • Diaz-Villasenor A, Burns AL, Hiriart M, Cebrian ME, Ostrosky-Wegman P: Arsenic-induced alteration in the expression of genes related to Type 2 diabetes mellitus. Toxicol. Appl. Pharmacol.225,123–133 (2007).
    • Tseng CH: Cardiovascular disease in arsenic-exposed subjects living in the arseniasis-hyperendemic areas in Taiwan. Atherosclerosis199(1),12–18 (2008).
    • 10  Guha Mazumder DN: Arsenic and liver disease. J. Indian Med. Assoc.99,311–314, 318–320 (2001).
    • 11  Wang ZY, Chen Z: Acute promyelocytic leukemia: from highly fatal to highly curable. Blood111,2505–2515 (2008).
    • 12  Dilda PJ, Hogg PJ: Arsenical-based cancer drugs. Cancer Treat. Rev.33,542–564 (2007).
    • 13  Westervelt P, Brown RA, Adkins DR et al.: Sudden death among patients with acute promyelocytic leukemia treated with arsenic trioxide. Blood98,266–271 (2001).
    • 14  Bencko V, Dobisova A, Macaj M: Arsenic in the hair of a non-occupationally exposed population. Atmos. Environ.5,275–279 (1971).
    • 15  Zakharyan RA, Sampayo-Reyes A, Healy SM et al.: Human monomethylarsonic acid (MMA[V]) reductase is a member of the glutathione-S-transferase superfamily. Chem. Res. Toxicol.14,1051–1057 (2001).
    • 16  Lin S, Shi Q, Nix FB et al.: A novel S-adenosyl-L-methionine:arsenic(III) methyltransferase from rat liver cytosol. J. Biol. Chem.277,10795–10803 (2002).
    • 17  Zakharyan RA, Ayala-Fierro F, Cullen WR, Carter DM, Aposhian HV: Enzymatic methylation of arsenic compounds. VII. Monomethylarsonous acid (MMAIII) is the substrate for MMA methyltransferase of rabbit liver and human hepatocytes. Toxicol. Appl. Pharmacol.158,9–15 (1999).
    • 18  Vahter M, Lind B: Concentrations of arsenic in urine of the general population in Sweden. Sci. Total Environ.54,1–12 (1986).
    • 19  Johnson LR, Farmer JG: Use of human metabolic studies and urinary arsenic speciation in assessing arsenic exposure. Bull. Environ. Contam. Toxicol.46,53–61 (1991).
    • 20  Hall M, Chen Y, Ahsan H et al.: Blood arsenic as a biomarker of arsenic exposure: results from a prospective study. Toxicology225,225–233 (2006).
    • 21  Rossman TG: Mechanism of arsenic carcinogenesis: an integrated approach. Mutat. Res.533,37–65 (2003).•• Interesting review that tries to integrate arsenic metabolism, genotoxic and nongenotoxic mechanisms and incidence of arsenic-related cancer. In contrast to the previous lack of good animal models to explain arsenic-related mechanisms, the paper reviews some promising new models for these cancers. The importance of the metabolism of inorganic arsenic to various methylated forms is also included.
    • 22  Vahter M, Concha G: Role of metabolism in arsenic toxicity. Pharmacol. Toxicol.89,1–5 (2001).
    • 23  Hsueh YM, Ko YF, Huang YK et al.: Determinants of inorganic arsenic methylation capability among residents of the Lanyang Basin, Taiwan: arsenic and selenium exposure and alcohol consumption. Toxicol. Lett.137,49–63 (2003).
    • 24  Vahter M: Methylation of inorganic arsenic in different mammalian species and population groups. Sci. Prog.82(Pt 1),69–88 (1999).
    • 25  Loffredo CA, Aposhian HV, Cebrian ME, Yamauchi H, Silbergeld EK: Variability in human metabolism of arsenic. Environ. Res.92,85–91 (2003).
    • 26  Chung JS, Kalman DA, Moore LE et al.: Family correlations of arsenic methylation patterns in children and parents exposed to high concentrations of arsenic in drinking water. Environ. Health Perspect.110,729–733 (2002).
    • 27  Wood TC, Salavagionne OE, Mukherjee B et al.: Human arsenic methyltransferase (AS3MT) pharmacogenetics: gene resequencing and functional genomics studies. J. Biol. Chem.281,7364–7373 (2006).
    • 28  Thomas DJ, Li J, Waters SB et al.: Arsenic (+3 oxidation state) methyltransferase and the methylation of arsenicals. Exp. Biol. Med. (Maywood)232,3–13 (2007).
    • 29  Cullen WR, Reimer KJ: Arsenic speciation in the environment. Chem. Rev.89,713–764 (1989).•• Together with [30], is very important in the context of this review and in the field assessing for the biological effects of arsenic. They represent two important steps in the understanding of the arsenic metabolism pathways, since they proposed the two models usually used to explain how inorganic arsenic is metabolized in the body.
    • 30  Hayakawa T, Kobayashi Y, Cui X, Hirano S: A new metabolic pathway of arsenite: arsenic–glutathione complexes are substrates for human arsenic methyltransferase Cyt19. Arch. Toxicol.79,183–191 (2005).•• Together with [29], is very important in the context of this review and in the field assessing for the biological effects of arsenic. They represent two important steps in the understanding of the arsenic metabolism pathways, since they proposed the two models usually used to explain how inorganic arsenic is metabolized in the body.
    • 31  Naranmandura H, Suzuki N, Suzuki KT: Trivalent arsenicals are bound to proteins during reductive methylation. Chem. Res. Toxicol.19,1010–1018 (2006).
    • 32  Meza MM, Yu L, Rodriguez YY et al.: Developmentally restricted genetic determinants of human arsenic metabolism: association between urinary methylated arsenic and CYT19 polymorphisms in children. Environ. Health Perspect.113,775–781 (2005).
    • 33  Schläwicke Engstrom K, Broberg K, Concha G, Nermell B, Warholm M, Vahter M: Genetic polymorphisms influencing arsenic metabolism: evidence from Argentina. Environ. Health Perspect.115,599–605 (2007).
    • 34  Lindberg AL, Kumar R, Goessler W et al.: Metabolism of low-dose inorganic arsenic in a central European population: influence of sex and genetic polymorphisms. Environ. Health Perspect.115,1081–1086 (2007).
    • 35  Hernández A, Xamena N, Surralles J et al.: Role of the Met(287)Thr polymorphism in the AS3MT gene on the metabolic arsenic profile. Mutat. Res.637,80–92 (2008).
    • 36  Hernández A, Xamena N, Sekaran C et al.: High arsenic metabolic efficiency in AS3MT287Thr allele carriers. Pharmacogenet. Genomics18,349–355 (2008).
    • 37  de Chaudhuri S, Ghosh P, Sarma N et al.: Genetic variants associated with arsenic susceptibility: study of purine nucleoside phosphorylase, arsenic (+3) methyltransferase, and glutathione S-transferase Ω genes. Environ. Health Perspect.116,501–505 (2008).
    • 38  Fujihara J, Kunito T, Takeshita H: Frequency of two human glutathione-S-transferase omega-1 polymorphisms (E155 deletion and E208K) in Ovambo and Japanese populations using the PCR-based genotyping method. Clin. Chem. Lab. Med.45,621–624 (2007).
    • 39  Radabaugh TR, Sampayo-Reyes A, Zakharyan RA, Aposhian HV: Arsenate reductase II. Purine nucleoside phosphorylase in the presence of dihydrolipoic acid is a route for reduction of arsenate to arsenite in mammalian systems. Chem. Res. Toxicol.15,692–698 (2002).
    • 40  Gregus Z, Nemeti B: Purine nucleoside phosphorylase as a cytosolic arsenate reductase. Toxicol. Sci.70,13–19 (2002).
    • 41  Yu L, Kalla K, Guthrie E, Vidrine A, Klimecki WT: Genetic variation in genes associated with arsenic metabolism: glutathione S-transferase Ω 1-1 and purine nucleoside phosphorylase polymorphisms in European and indigenous Americans. Environ. Health Perspect.111,1421–1427 (2003).
    • 42  Nemeti B, Csanaky I, Gregus Z: Arsenate reduction in human erythrocytes and rats – testing the role of purine nucleoside phosphorylase. Toxicol. Sci.74,22–31 (2003).
    • 43  Patterson TJ, Ngo M, Aronov PA, Reznikova TV, Green PG, Rice RH: Biological activity of inorganic arsenic and antimony reflects oxidation state in cultured human keratinocytes. Chem. Res. Toxicol.16,1624–1631 (2003).
    • 44  Nemeti B, Gregus Z: Glutathione-dependent reduction of arsenate in human erythrocytes – a process independent of purine nucleoside phosphorylase. Toxicol. Sci.82,419–428 (2004).
    • 45  Whitbread AK, Tetlow N, Eyre HJ, Sutherland GR, Board PG: Characterization of the human Ω class glutathione transferase genes and associated polymorphisms. Pharmacogenetics13,131–144 (2003).
    • 46  McCollum G, Keng PC, States JC, McCabe MJ Jr: Arsenite delays progression through each cell cycle phase and induces apoptosis following G2/M arrest in U937 myeloid leukemia cells. J. Pharmacol. Exp. Ther.313,877–887 (2005).
    • 47  Marnell LL, Garcia-Vargas GG, Chowdhury UK et al.: Polymorphisms in the human monomethylarsonic acid (MMAV) reductase/hGSTO1 gene and changes in urinary arsenic profiles. Chem. Res. Toxicol.16,1507–1513 (2003).
    • 48  Mukherjee B, Salavaggione OE, Pelleymounter LL et al.: Glutathione S-transferase Ω 1 and Ω 2 pharmacogenomics. Drug Metab. Dispos.34,1237–1246 (2006).
    • 49  Tanaka-Kagawa T, Jinno H, Hasegawa T et al.: Functional characterization of two variant human GSTO 1-1s (Ala140Asp and Thr217Asn). Biochem. Biophys. Res. Commun.301,516–520 (2003).
    • 50  Schmuck EM, Board PG, Whitbread AK et al.: Characterization of the monomethylarsonate reductase and dehydroascorbate reductase activities of Ω class glutathione transferase variants: implications for arsenic metabolism and the age-at-onset of Alzheimer’s and Parkinson’s diseases. Pharmacogenet. Genomics15,493–501 (2005).
    • 51  Paiva L, Marcos R, Creus A, Coggan M, Oakley AJ, Board PG: Polymorphism of glutathione transferase Ω 1 in a population exposed to a high environmental arsenic burden. Pharmacogenet. Genomics18,1–10 (2008).
    • 52  Board PG, Coggan M, Chelvanayagam G et al.: Identification, characterization, and crystal structure of the Ω class glutathione transferases. J. Biol. Chem.275,24798–24806 (2000).
    • 53  Ahsan H, Chen Y, Kibriya MG et al.: Arsenic metabolism, genetic susceptibility, and risk of premalignant skin lesions in Bangladesh. Cancer Epidemiol. Biomarkers Prev.16,1270–1278 (2007).• Large case–control study to investigate interindividual variability in susceptibility to health effects of inorganic arsenic due to arsenic metabolism efficiency, genetic factors and their interaction. A total of 594 cases of arsenic-induced skin lesions and 1041 controls were included in the study. GSTO1 and MTHFR polymorphisms were selected. The analysis of joint effects of genotypes/diplotypes with water arsenic concentration and urinary %monomethylarsonic suggests additivity of these factors.
    • 54  Whitbread AK, Masoumi A, Tetlow N, Schmuck E, Coggan M, Board PG: Characterization of the Ω class of glutathione transferases. Meth. Enzymol.401,78–99 (2005).
    • 55  Chowdhury UK, Zakharyan RA, Avram MD et al.: Inorganic arsenic biotransformation and MMA(V) reductase/GSTO1-1 knockout mice. Presented at: 44th Annual Meeting of the Society of Toxicology, New Orleans, LA, USA, 6–10 March (2005).
    • 56  Chowdhury UK, Zakharyan RA, Hernandez A, Avram MD, Kopplin MJ, Aposhian HV: Glutathione-S-transferase-omega [MMA(V) reductase] knockout mice: enzyme and arsenic species concentrations in tissues after arsenate administration. Toxicol. Appl. Pharmacol.216,446–457 (2006).
    • 57  Flora SJ, Bhadauria S, Kannan GM, Singh N: Arsenic induced oxidative stress and the role of antioxidant supplementation during chelation: a review. J. Environ. Biol.28,333–347 (2007).
    • 58  Pi J, Yamauchi H, Kumagai Y et al.: Evidence for induction of oxidative stress caused by chronic exposure of Chinese residents to arsenic contained in drinking water. Environ. Health Perspect.110,331–336 (2002).
    • 59  Fujino Y, Guo X, Liu J et al.: Chronic arsenic exposure and urinary 8-hydroxy-2´-deoxyguanosine in an arsenic-affected area in Inner Mongolia, China. J. Expo. Anal. Environ. Epidemiol.15,147–152 (2005).
    • 60  Wang TS, Shu YF, Liu YC, Jan KY, Huang H: Glutathione peroxidase and catalase modulate the genotoxicity of arsenite. Toxicology121,229–237 (1997).
    • 61  Schuliga M, Chouchane S, Snow ET: Upregulation of glutathione-related genes and enzyme activities in cultured human cells by sublethal concentrations of inorganic arsenic. Toxicol. Sci.70,183–192 (2002).
    • 62  Cotton SC, Sharp L, Little J, Brockton N: Glutathione S-transferase polymorphisms and colorectal cancer: a HuGE review. Am. J. Epidemiol.151,7–32 (2000).
    • 63  Strange RC, Lear JT, Fryer AA: Polymorphism in glutathione S-transferase loci as a risk factor for common cancers. Arch. Toxicol.20(Suppl.),419–428 (1998).
    • 64  Tew KD: Glutathione-associated enzymes in anticancer drug resistance. Cancer Res.54,4313–4320 (1994).
    • 65  Harries LW, Stubbins MJ, Forman D, Howard GC, Wolf CR: Identification of genetic polymorphisms at the glutathione S-transferase Pi locus and association with susceptibility to bladder, testicular and prostate cancer. Carcinogenesis18,641–644 (1997).
    • 66  Kile ML, Houseman EA, Rodrigues E et al.: Toenail arsenic concentrations, GSTT1 gene polymorphisms, and arsenic exposure from drinking water. Cancer Epidemiol. Biomarkers Prev.14,2419–2426 (2005).
    • 67  Bernardini S, Nuccetelli M, Noguera NI et al.: Role of GSTP1-1 in mediating the effect of As2O3 in the acute promyelocytic leukemia cell line NB4. Ann. Hematol.85,681–687 (2006).
    • 68  McCarty KM, Chen YC, Quamruzzaman Q et al.: Arsenic methylation, GSTT1, GSTM1, GSTP1 polymorphisms, and skin lesions. Environ. Health Perspect.115,341–345 (2007).
    • 69  Breton CV, Kile ML, Catalano PJ et al.: GSTM1 and APE1 genotypes affect arsenic-induced oxidative stress: a repeated measures study. Environ. Health6,39 (2007).
    • 70  Chiou HY, Hsueh YM, Hsieh LL et al.: Arsenic methylation capacity, body retention, and null genotypes of glutathione S-transferase M1 and T1 among current arsenic-exposed residents in Taiwan. Mutat. Res.386,197–207 (1997).
    • 71  Steinmaus C, Moore LE, Shipp M et al.: Genetic polymorphisms in MTHFR 677 and 1298, GSTM1 and T1, and metabolism of arsenic. J. Toxicol. Environ. Health A.70,159–170 (2007).
    • 72  Marcos R, Martinez V, Hernandez A et al.: Metabolic profile in workers occupationally exposed to arsenic: role of GST polymorphisms. J. Occup. Environ. Med.48,334–341 (2006).
    • 73  Ghosh P, Basu A, Mahata J et al.: Cytogenetic damage and genetic variants in the individuals susceptible to arsenic-induced cancer through drinking water. Int. J. Cancer118,2470–2478 (2006).
    • 74  Lin GF, Du H, Chen JG et al.: Glutathione S-transferases M1 and T1 polymorphisms and arsenic content in hair and urine in two ethnic clans exposed to indoor combustion of high arsenic coal in southwest Guizhou, China. Arch. Toxicol.81,545–551 (2007).
    • 75  Adonis M, Martinez V, Marin P, Berrios D, Gil L: Smoking habit and genetic factors associated with lung cancer in a population highly exposed to arsenic. Toxicol. Lett.159,32–37 (2005).
    • 76  Hsu LI, Chiu AW, Huan SK et al.: SNPs of GSTM1,T1,P1, epoxide hydrolase and DNA repair enzyme XRCC1 and risk of urinary transitional cell carcinoma in southwestern Taiwan. Toxicol. Appl. Pharmacol.228,144–155 (2008).
    • 77  Wang YH, Wu MM, Hong CT et al.: Effects of arsenic exposure and genetic polymorphisms of p53, glutathione S-transferase M1, T1, and P1 on the risk of carotid atherosclerosis in Taiwan. Atherosclerosis192,305–312 (2007).
    • 78  McCarty KM, Ryan L, Houseman EA et al.: A case–control study of GST polymorphisms and arsenic related skin lesions. Environ. Health6,5 (2007).
    • 79  Chen YC, Xu L, Guo YL et al.: Polymorphisms in GSTT1 and p53 and urinary transitional cell carcinoma in south-western Taiwan: a preliminary study. Biomarkers9,386–394 (2004).
    • 80  Lin GF, Du H, Chen JG et al.: Arsenic-related skin lesions and glutathione S-transferase P1 A1578G (Ile105Val) polymorphism in two ethnic clans exposed to indoor combustion of high arsenic coal in one village. Pharmacogenet. Genomics16,863–871 (2006).
    • 81  Marafante E, Vahter M, Envall J: The role of the methylation in the detoxication of arsenate in the rabbit. Chem. Biol. Interact.56,225–238 (1985).
    • 82  Marafante E, Vahter M: The effect of methyltransferase inhibition on the metabolism of [74As]arsenite in mice and rabbits. Chem. Biol. Interact.50,49–57 (1984).
    • 83  Vahter M, Marafante E: Effects of low dietary intake of methionine, choline or proteins on the biotransformation of arsenite in the rabbit. Toxicol. Lett.37,41–46 (1987).
    • 84  Nelson GM, Ahlborn GJ, Delker DA et al.: Folate deficiency enhances arsenic effects on expression of genes involved in epidermal differentiation in transgenic K6/ODC mouse skin. Toxicology241,134–145 (2007).
    • 85  Frosst P, Blom HJ, Milos R et al.: A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat. Genet.10,111–113 (1995).
    • 86  Gamble MV, Ahsan H, Liu X et al.: Folate and cobalamin deficiencies and hyperhomocysteinemia in Bangladesh. Am. J. Clin. Nutr.81,1372–1377 (2005).
    • 87  Gamble MV, Liu X, Ahsan H et al.: Folate and arsenic metabolism: a double-blind, placebo-controlled folic acid-supplementation trial in Bangladesh. Am. J. Clin. Nutr.84,1093–1101 (2006).
    • 88  Gamble MV, Liu X, Ahsan H et al.: Folate, homocysteine, and arsenic metabolism in arsenic-exposed individuals in Bangladesh. Environ. Health Perspect.113,1683–1688 (2005).•• States the possible modulation of the nutritional status on the effects of arsenic. This cross-sectional study characterizes the relationships among folate, cobalamin, homocysteine and arsenic metabolism in Bangladeshi adults. Collectively, the reported data suggest that folate and other factors involved in one-carbon metabolism influence arsenic methylation.
    • 89  Gamble MV, Liu X, Slavkovich V et al.: Folic acid supplementation lowers blood arsenic. Am. J. Clin. Nutr.86,1202–1209 (2007).
    • 90  Gebel TW: Genotoxicity of arsenical compounds. Int. J. Hyg. Environ. Health.203,249–262 (2001).
    • 91  Bau DT, Wang TS, Chung CH, Wang AS, Wang AS, Jan KY: Oxidative DNA adducts and DNA-protein cross-links are the major DNA lesions induced by arsenite. Environ. Health Perspect.110(Suppl. 5),753–756 (2002).
    • 92  Hartwig A, Pelzer A, Asmuss M, Burkle A: Very low concentrations of arsenite suppress poly(ADP-ribosyl)ation in mammalian cells. Int. J. Cancer104,1–6 (2003).
    • 93  Poonepalli A, Balakrishnan L, Khaw AK et al.: Lack of poly(ADP-ribose) polymerase-1 gene product enhances cellular sensitivity to arsenite. Cancer Res.65,10977–10983 (2005).
    • 94  Schwerdtle T, Walter I, Hartwig A: Arsenite and its biomethylated metabolites interfere with the formation and repair of stable BPDE-induced DNA adducts in human cells and impair XPAzf and Fpg. DNA Repair (Amst.)2,1449–1463 (2003).
    • 95  Andrew AS, Karagas MR, Hamilton JW: Decreased DNA repair gene expression among individuals exposed to arsenic in United States drinking water. Int. J. Cancer104,263–268 (2003).• Interesting paper that is the only one that has studied the expression of critical members of the nucleotide-excision repair complex in human populations exposed to arsenic. Found that ERCC1, XPF and XPB expression was inversely correlated with the exposure measured by toenail arsenic levels.
    • 96  Breton CV, Zhou W, Kile ML et al.: Susceptibility to arsenic-induced skin lesions from polymorphisms in base excision repair genes. Carcinogenesis28,1520–1525 (2007).
    • 97  Banerjee M, Sarkar J, Das JK et al.: Polymorphism in the ERCC2 codon 751 is associated with arsenic-induced premalignant hyperkeratosis and significant chromosome aberrations. Carcinogenesis28,672–676 (2007).
    • 98  McCarty KM, Smith TJ, Zhou W et al.: Polymorphisms in XPD (Asp312Asn and Lys751Gln) genes, sunburn and arsenic-related skin lesions. Carcinogenesis28,1697–1702 (2007).
    • 99  Ahsan H, Chen Y, Wang Q, Slavkovich V, Graziano JH, Santella RM: DNA repair gene XPD and susceptibility to arsenic-induced hyperkeratosis. Toxicol. Lett.143,123–131 (2003).
    • 100  Applebaum KM, Karagas MR, Hunter DJ et al.: Polymorphisms in nucleotide excision repair genes, arsenic exposure, and non-melanoma skin cancer in New Hampshire. Environ. Health Perspect.115,1231–1236 (2007).
    • 101  Spitz MR, Wu X, Wang Y et al.: Modulation of nucleotide excision repair capacity by XPD polymorphisms in lung cancer patients. Cancer Res.61,1354–1357 (2001).
    • 102  Mass MJ, Wang L: Arsenic alters cytosine methylation patterns of the promoter of the tumor suppressor gene p53 in human lung cells: a model for a mechanism of carcinogenesis. Mutat. Res.386,263–277 (1997).
    • 103  Boonchai W, Walsh M, Cummings M, Chenevix-Trench G: Expression of p53 in arsenic-related and sporadic basal cell carcinoma. Arch. Dermatol.136,195–198 (2000).
    • 104  Hsu CH, Yang SA, Wang JY, Yu HS, Lin SR: Mutational spectrum of p53 gene in arsenic-related skin cancers from the blackfoot disease endemic area of Taiwan. Br. J. Cancer.80,1080–1086 (1999).
    • 105  de Chaudhuri S, Mahata J, Das JK et al.: Association of specific p53 polymorphisms with keratosis in individuals exposed to arsenic through drinking water in West Bengal, India. Mutat. Res.601,102–112 (2006).
    • 106  Chen YC, Guo YL, Su HJ et al.: Arsenic methylation and skin cancer risk in southwestern Taiwan. J. Occup. Environ. Med.45,241–248 (2003).
    • 107  Kessel M, Liu SX, Xu A, Santella R, Hei TK: Arsenic induces oxidative DNA damage in mammalian cells. Mol. Cell. Biochem.234–235(1–2),301–308 (2002).
    • 108  Lee TC, Ho IC: Modulation of cellular antioxidant defense activities by sodium arsenite in human fibroblasts. Arch. Toxicol.69,498–504 (1995).
    • 109  Nandi D, Patra RC, Swarup D: Oxidative stress indices and plasma biochemical parameters during oral exposure to arsenic in rats. Food Chem. Toxicol.44,1579–1584 (2006).
    • 110  Mishra D, Mehta A, Flora SJ: Reversal of arsenic-induced hepatic apoptosis with combined administration of DMSA and its analogues in guinea pigs: role of glutathione and linked enzymes. Chem. Res. Toxicol.21,400–407 (2008).
    • 111  Xue DB, Zhang WH, Yun XG et al.: Regulating effects of arsenic trioxide on cell death pathways and inflammatory reactions of pancreatic acinar cells in rats. Chin. Med. J. (Engl.)120,690–695 (2007).
    • 112  Hsueh YM, Lin P, Chen HW et al.: Genetic polymorphisms of oxidative and antioxidant enzymes and arsenic-related hypertension. J. Toxicol. Environ. Health A.68,1471–1484 (2005).
    • 113  Ahsan H, Chen Y, Kibriya MG et al.: Susceptibility to arsenic-induced hyperkeratosis and oxidative stress genes myeloperoxidase and catalase. Cancer Lett.201,57–65 (2003).
    • 114  Falkner KC, McCallum GP, Cherian MG, Bend JR: Effects of acute sodium arsenite administration on the pulmonary chemical metabolizing enzymes, cytochrome P-450 monooxygenase, NAD(P)H:quinone acceptor oxidoreductase and glutathione S-transferase in guinea pig: comparison with effects in liver and kidney. Chem. Biol. Interact.86,51–68 (1993).
    • 115  Jacobs JM, Nichols CE, Andrew AS et al.: Effect of arsenite on induction of CYP1A, CYP2B, and CYP3A in primary cultures of rat hepatocytes. Toxicol. Appl. Pharmacol.157,51–59 (1999).
    • 116  Simeonova PP, Luster MI: Arsenic and atherosclerosis. Toxicol. Appl. Pharmacol.198,444–449 (2004).
    • 117  Hsieh YC, Hsieh FI, Lien LM, Chou YL, Chiou HY, Chen CJ: Risk of carotid atherosclerosis associated with genetic polymorphisms of apolipoprotein E and inflammatory genes among arsenic exposed residents in Taiwan. Toxicol. Appl. Pharmacol.227,1–7 (2008).
    • 118  Harrington JR: The role of MCP-1 in atherosclerosis. Stem Cells18,65–66 (2000).
    • 119  Simeonova PP, Hulderman T, Harki D, Luster MI: Arsenic exposure accelerates atherogenesis in apolipoprotein E(-/-) mice. Environ. Health Perspect.111,1744–1748 (2003).
    • 120  Liu Z, Styblo M, Rosen BP: Methylarsonous acid transport by aquaglyceroporins. Environ. Health Perspect.114,527–531 (2006).•• Addresses how methylated arsenicals pass through cell membranes. Demonstrates that aquaglyceroporins differ both in selectivity for and transport rates of trivalent arsenicals into the cells. This study, together with others, leads us to suggest a new set of genes (polymorphisms) to be used in future association studies.
    • 121  Liu Z, Carbrey JM, Agre P, Rosen BP: Arsenic trioxide uptake by human and rat aquaglyceroporins. Biochem. Biophys. Res. Commun.316,1178–1185 (2004).
    • 122  Liu Z, Sanchez MA, Jiang X, Boles E, Landfear SM, Rosen BP: Mammalian glucose permease GLUT1 facilitates transport of arsenic trioxide and methylarsonous acid. Biochem. Biophys. Res. Commun.351,424–430 (2006).
    • 123  Thomas DJ: Molecular processes in cellular arsenic metabolism. Toxicol. Appl. Pharmacol.222,365–373 (2007).•• Provides a basis for understanding the modes of action by which inorganic arsenic acts as a toxin and a carcinogen. Proposes the development of biologically based dose–response models describing the toxic and carcinogenic actions of arsenicals.