Many inorganics are human carcinogens and pose major hazards to after environmental or occupational exposure. Arsenic and cadmium are two of the most important inorganic carcinogens and with arsenic alone over 100 million people world-wide are exposed to clearly unhealthy levels via contaminated drinking water in what is likely the worst mass poisoning in human history. Defining carcinogenic mechanisms will greatly aid in designing prevention or intervention strategies and in assigning appropriate levels of risk to these exposures. The primary goal of the ICS is to define the molecular mechanisms of carcinogenic action of arsenic and cadmium under the project titled Molecular Mechanisms of Inorganic Carcinogenesis. Since they can impact carcinogenicity and potentially provide a means for prevention or intervention, mechanisms of adaptation and acquired tolerance are studied as well. Emphasis is placed on key factors that dictate sensitivity, such as early-life exposure and poor expression of critical adaptive genes. These inorganics attack various targets sites in humans. Arsenic causes skin, urinary bladder, lung, kidney, liver, and prostatic malignancies and likely uterine cancers. Cadmium is primarily associated with human lung, prostatic and kidney cancers and recently pancreatic cancers. All these sites may have distinct differences in mode of action and adaption. Thus, various in vitro and in vivo model systems have been developed to study important molecular targets in targets tissues, with an emphasis on human relevance. These inorganic carcinogens likely have multiple mechanisms that are site and cell specific. A reproducible rodent model where inorganic arsenic acts a complete carcinogen has been developed in which brief in utero arsenic exposure in mice leads to tumors or proliferative lesions of the urogenital system, liver, lung and adrenal in the offspring as adults. The urogenital system lesions include transplacental arsenic-induced or initiated tumors of the ovaries, uterus, vagina, and bladder and proliferative lesions of the kidney. These results are in accord with human studies that indicate the liver, urinary bladder, lung, kidney and uterus are target tissues of arsenic carcinogenesis. Molecular mechanism studies indicate disruption of estrogen signaling by in utero exposure to arsenic contributes to the liver, lung and urogenital system malignancies, in part through aberrant activation of estrogen receptor-alpha. Indeed, we find that tumors and proliferative lesions of the urogenital system, including the uterus, ovary, vagina and urinary bladder, are greatly enhanced by postnatal exposure to synthetic estrogens like diethylstilbestrol. We also find evidence of aberrant estrogen signaling in arsenic exposed human liver. Further molecular characterization of arsenic-induced in utero tumor initiation is underway, including aberrant gene imprinting, using this model of arsenic carcinogenesis. We hypothesize that arsenic in utero may attack a critical pool of stem cells in target organs and induces aberrant genetic reprograming as part of its carcinogenic mechanism. These studies have important public health implications, including the identification of early life period as a time of very high sensitivity to arsenic. Human data is now emerging that fetal and/or early life exposure to arsenic is clearly carcinogenic. Further study will include prenatal arsenic exposure combined with exposures to urinary bladder and renal tumor promoters in mice to enhance the carcinogenic response to arsenic in these key human target organs. Various in vitro cell transformation model systems have also been developed to study inorganic carcinogenesis. In doing these studies we select cells with relevance to the human targets of arsenic, cadmium or lead carcinogenesis, and use low-level exposures for long periods, which approximates typical human exposures and avoids supra-physiological responses associated with acute high doses that could have limited relevance to the carcinogenic process. A human prostate epithelial cell line has been malignantly transformed with cadmium and arsenic, both potential human prostatic carcinogens. Additional work indicates the arsenic and cadmium transformants both acquire androgen independence, an event associated with a very poor clinical prognosis in prostatic cancer patients, largely through androgen receptor by-pass related mechanisms. Molecular dissection of the events associated with arsenic- or cadmium-induced malignant transformation in this and other human cell lines will continue with a focus of aberrant expression of genes critical to the carcinogenic process. In addition, a human prostate stem cell line has been developed and will be studied as a potential target cell population of these carcinogenic inorganics, and it clearly shows a survival selection advantage, a tleast for arsenic, along its was to malignant transformation. Furthermore, we have successfully transformed a human pancreatic ductal cell with cadmium, which fortifies a possible role of cadmium in this deadly disease. Similarly, arsenic has induced malignant transformation of human skin keratinocytes. The study of this arsenic-induced skin cancer model indicates that it occurs through a very different mechanism from internal cancers, one which involves apoptotic by-pass and aberrant survival of damaged skin cells with a strong possibility of survival selection of skin stem cells. The latter is now being tested in various model systems. Many cell biomethylate arsenic using specific methyltransferases S-adenosylmethionine (SAM) as the methyl group donor, which could compete for cellular methyl groups used in other enzymatic methylation reactions, such as DNA methylation. Altered DNA methylation is a common epigenetic finding in cancer, but we also find a loss of DNA methylation in arsenic target cells that do not biomethylate the metalloid. In this regard, after protracted low level arsenic exposure, the normal human prostate epithelial cells acquires a malignant phenotype with DNA hypomethylation, indicative of disrupted methyl metabolism, and shows arsenic adaptation involving glutathione overproduction and enhanced arsenic efflux. Thus, the interplay between methyl and glutathione metabolism during this progressive arsenic adaptation was studied. Arsenic-treated cells showed a time-dependent increase in adaption to arsenic toxicity and a marked increase in homocysteine (Hcy) levels. A marked suppression of SAM levels occurred with decreased methionine adenosyltransferase 2A (converts methionine to SAM) expression and increased negative regulator methionine adenosyltransferase B, suggesting reduced conversion of Hcy to SAM. Consistent with Hcy overproduction, activity of S-adenosylhomocysteine hydrolase (converts S-adenosylhomocysteine to Hcy) was increased. Cystathionine beta-synthase, a key gene in the transsulfuration pathway, and various glutathione production genes were increased, resulting greatly increased glutathione. Arsenic efflux increased along with expression of ATP-binding cassette protein C1, which effluxes arsenic as a glutathione conjugate. Genomic DNA hypomethylation was observed with arsenic exposure, indicating that the disruption in methyl metabolism h [summary truncated at 7800 characters]

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Intramural Research (Z01)
Project #
1Z01BC005488-23
Application #
7732881
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
23
Fiscal Year
2008
Total Cost
$1,786,280
Indirect Cost
Name
National Cancer Institute Division of Basic Sciences
Department
Type
DUNS #
City
State
Country
United States
Zip Code
Pelch, Katherine E; Tokar, Erik J; Merrick, B Alex et al. (2015) Differential DNA methylation profile of key genes in malignant prostate epithelial cells transformed by inorganic arsenic or cadmium. Toxicol Appl Pharmacol 286:159-67
Tokar, Erik J; Diwan, Bhalchandra A; Waalkes, Michael P (2010) Early life inorganic lead exposure induces testicular teratoma and renal and urinary bladder preneoplasia in adult metallothionein-knockout mice but not in wild type mice. Toxicology 276:5-10
Sun, Yang; Pi, Jingbo; Wang, Xueqian et al. (2009) Aberrant cytokeratin expression during arsenic-induced acquired malignant phenotype in human HaCaT keratinocytes consistent with epidermal carcinogenesis. Toxicology 262:162-70
Shimada, Hideaki; Narumi, Rika; Nagano, Masaaki et al. (2009) Strain difference of cadmium-induced testicular toxicity in inbred Wistar-Imamichi and Fischer 344 rats. Arch Toxicol 83:647-52
Waalkes, Michael P; Liu, Jie; Germolec, Dori R et al. (2008) Arsenic exposure in utero exacerbates skin cancer response in adulthood with contemporaneous distortion of tumor stem cell dynamics. Cancer Res 68:8278-85
Waalkes, Michael P; Liu, Jie; Diwan, Bhalchandra A (2007) Transplacental arsenic carcinogenesis in mice. Toxicol Appl Pharmacol 222:271-80
Pi, Jingbo; Bai, Yushi; Reece, Jeffrey M et al. (2007) Molecular mechanism of human Nrf2 activation and degradation: role of sequential phosphorylation by protein kinase CK2. Free Radic Biol Med 42:1797-806
Benbrahim-Tallaa, Lamia; Liu, Jie; Webber, Mukta M et al. (2007) Estrogen signaling and disruption of androgen metabolism in acquired androgen-independence during cadmium carcinogenesis in human prostate epithelial cells. Prostate 67:135-45
Benbrahim-Tallaa, Lamia; Webber, Mukta M; Waalkes, Michael P (2007) Mechanisms of acquired androgen independence during arsenic-induced malignant transformation of human prostate epithelial cells. Environ Health Perspect 115:243-7
Xie, Yaxiong; Liu, Jie; Benbrahim-Tallaa, Lamia et al. (2007) Aberrant DNA methylation and gene expression in livers of newborn mice transplacentally exposed to a hepatocarcinogenic dose of inorganic arsenic. Toxicology 236:7-15

Showing the most recent 10 out of 79 publications