Transplacental exposure to inorganic arsenic at a hepatocarcinogenic dose induces fetal gene expression changes in mice indicative of aberrant estrogen signaling and disrupted steroid metabolism

https://doi.org/10.1016/j.taap.2007.01.018Get rights and content

Abstract

Exposure to inorganic arsenic in utero in C3H mice produces hepatocellular carcinoma in male offspring when they reach adulthood. To help define the molecular events associated with the fetal onset of arsenic hepatocarcinogenesis, pregnant C3H mice were given drinking water containing 0 (control) or 85 ppm arsenic from day 8 to 18 of gestation. At the end of the arsenic exposure period, male fetal livers were removed and RNA isolated for microarray analysis using 22K oligo chips. Arsenic exposure in utero produced significant (p < 0.001) alterations in expression of 187 genes, with approximately 25% of aberrantly expressed genes related to either estrogen signaling or steroid metabolism. Real-time RT-PCR on selected genes confirmed these changes. Various genes controlled by estrogen, including X-inactive-specific transcript, anterior gradient-2, trefoil factor-1, CRP-ductin, ghrelin, and small proline-rich protein-2A, were dramatically over-expressed. Estrogen-regulated genes including cytokeratin 1–19 and Cyp2a4 were over-expressed, although Cyp3a25 was suppressed. Several genes involved with steroid metabolism also showed remarkable expression changes, including increased expression of 17β-hydroxysteroid dehydrogenase-7 (HSD17β7; involved in estradiol production) and decreased expression of HSD17β5 (involved in testosterone production). The expression of key genes important in methionine metabolism, such as methionine adenosyltransferase-1a, betaine-homocysteine methyltransferase and thioether S-methyltransferase, were suppressed. Thus, exposure of mouse fetus to inorganic arsenic during a critical period in development significantly alters the expression of various genes encoding estrogen signaling and steroid or methionine metabolism. These alterations could disrupt genetic programming at the very early life stage, which could impact tumor formation much later in adulthood.

Introduction

Inorganic arsenic is a human carcinogen, associated with tumors of the skin, urinary bladder, lung, liver, prostate, kidney, and possibly other sites (NRC, 2001, Morales et al., 2000, Centeno et al., 2002, IARC, 2004). We have shown that short-term exposure in mice to inorganic arsenic in utero produces a variety of internal tumors in the offspring when they reach adulthood (Waalkes et al., 2003, Waalkes et al., 2004a, Waalkes et al., 2006a, Waalkes et al., 2006b). Gestation is a period of high sensitivity to chemical carcinogenesis in rodents and probably in humans (Anderson et al., 2000). Inorganic arsenic can readily cross the rodent and human placenta and enter the fetus (Concha et al., 1998, NRC, 2001). After in utero exposure to inorganic arsenic at carcinogenic doses, significant amounts of inorganic arsenic and its methylated metabolites (DMA and MMA) are detected in various mouse fetal tissues including the liver (Devesa et al., 2006). In arsenic-exposed human populations all life stages of exposure are involved (IARC, 2004). Thus, it is likely that significant in utero arsenic exposure occurs in human populations, and it is prudent to assume that the transplacental carcinogenic risks defined in rodents may predict similar effects in humans.

The liver is a major target organ of arsenic toxicity (Lu et al., 2001, Mazumder, 2005) and carcinogenesis in humans (Chen et al., 1997, Zhou et al., 2002, Centeno et al., 2002, Chen and Ahsan, 2004). In accord with human data, transplacental exposure to inorganic arsenic induced a marked, dose-related increase in hepatocellular tumors, including carcinoma, in adult male mice (Waalkes et al., 2003, Waalkes et al., 2004a, Waalkes et al., 2006b). Genomic analysis of liver samples taken at necropsy 1–2 years after gestational arsenic exposure alone or combined with postnatal exposure to 12-O-teradecanoyl phorbol-13-acetate (TPA) revealed a variety of hepatic genes to be aberrantly expressed in adulthood, including genes critical to the carcinogenic process (Liu et al., 2004, Liu et al., 2006a, Liu et al., 2006b). Although the expression changes in adult mouse liver are clearly associated with liver tumors, whether they are involved in cancer causation specifically by arsenic cannot be defined in fully developed tumors. Thus, genomic analysis of early molecular events following gestational arsenic exposure is clearly warranted.

The spectrum of tumors and/or proliferative lesions induced by in utero arsenic exposure, including tumors of liver, ovary, adrenal, uterus and oviduct, resembles the potential targets of carcinogenic estrogens (Waalkes et al., 2003, Waalkes et al., 2004a, Waalkes et al., 2006a, Waalkes et al., 2006b). This has led us to the hypothesis that arsenic could somehow produce estrogen-like effects, possibly through estrogen receptor-alpha (ER-α), as part of the mechanisms causing tumor formation (Waalkes et al., 2004b). Aberrant over-expression of ER-α is associated with a variety of human and rodent tumors (Fishman et al., 1995). Indeed, in livers and liver tumors from male mice exposed to arsenic in utero, the over-expression of the ER-α and estrogen-linked cyclin D1 is a prominent feature, and a feminized pattern of hepatic metabolic enzyme genes is evident (Waalkes et al., 2004b, Liu et al., 2004, Liu et al., 2006b). Samples of human livers from populations highly exposed to inorganic arsenic also show ER-α over-expression (Waalkes et al., 2004b).

Thus, this study investigated aberrant gene expression in the fetal male livers following in utero exposure to a hepatocarcinogenic dose of arsenic. Global genomic analysis was performed through the National Center for Toxicogenomics, using the Agilent 22K chip array. Expression of key genes was followed up by real-time RT-PCR analysis. This study clearly showed that in utero arsenic exposure produced dramatic alterations in gene expression in fetal liver, providing evidence for enhanced estrogen signaling and aberrant steroid metabolism in the developing fetus as a result of transplacental arsenic exposure. This arsenic-induced early life stage disruption of genetic programming could potentially lead to tumor formation much later in adulthood.

Section snippets

Chemicals

Sodium arsenite (NaAsO2) was obtained from Sigma Chemical Co. (St. Louis, MO) and dissolved in the drinking water at 85 mg arsenic/L (85 ppm). The Agilent 22K mouse oligo array was obtained from Agilent Technologies (Palo Alto, CA).

Animal treatment and sample collection

Timed pregnant C3H mice were given drinking water containing 85 ppm arsenic or unaltered water ad libitum from day 8 to day 18 of gestation. At day 18 of gestation, mice were killed by CO2 asphyxiation and fetuses removed. Only male fetal livers were used for the

Microarray analysis of aberrantly expressed genes

Total RNA from male fetal liver samples of control and arsenic-exposed mice was subjected to microarray analysis. Under the criteria of p < 0.001 by the Rosetta Resolver (v4.0) system, the transcript levels of 187 genes among 22,000 contained on the array were significantly altered by arsenic exposure compared to control. Clustering analysis of these altered genes is shown in Fig. 1. A cluster of increased genes (shown in red) included various genes related to estrogen signaling, and a cluster of

Discussion

The hypothesis that inorganic arsenic might somehow act through aberrant activation of estrogen signaling pathways (Waalkes et al., 2004b) comes from several lines of evidence. This includes the fact that the transplacental arsenic carcinogenesis shows consistent targets (i.e., liver, ovary, adrenal, uterus) which are also targets of broad range or tissue-selective carcinogenic estrogens (Birnbaum and Fenton, 2003, Newbold, 2004). In addition, estrogen-linked gene/protein over-expressions are

Acknowledgments

The authors thank Drs. Erik Tokar, Ronald Cannon and Larry Keefer for their critical review of the manuscript. Research was supported in part by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research, the Federal funds from the National Cancer Institute, National Institutes of Health, under contract No. NO1-CO-12400, and the National Center for Toxicogenomics at NIEHS. The content of this publication does not necessarily reflect the views or policies

References (48)

  • L. Thim et al.

    Isolation and characterization of putative trefoil peptide receptors

    Regul. Pept.

    (2000)
  • L. Vergnes et al.

    A cluster of eight hydroxysteroid dehydrogenase genes belonging to the aldo-keto reductase supergene family on mouse chromosome 13

    J. Lipid Res.

    (2003)
  • M.P. Waalkes et al.

    Transplacental carcinogenicity of inorganic arsenic in the drinking water: induction of hepatic, ovarian, pulmonary, and adrenal tumors in mice

    Toxicol. Appl. Pharmacol.

    (2003)
  • M.P. Waalkes et al.

    Tumors and proliferative lesions of liver, urinary bladder, adrenal, lung, and kidney in male CD1 mice after transplacental exposure to inorganic arsenic and postnatal treatment with diethylstilbestrol or tamoxifen

    Toxicol. Appl. Pharmacol.

    (2006)
  • L.M. Anderson et al.

    Critical windows of exposure for children's health: cancer in human epidemiological studies and neoplasms in experimental animal models

    Environ. Health Perspect.

    (2000)
  • L.S. Birnbaum et al.

    Cancer and developmental exposure to endocrine disruptors

    Environ. Health Perspect.

    (2003)
  • J.A. Centeno et al.

    Pathology related to chronic arsenic exposure

    Environ. Health Perspect.

    (2002)
  • Y. Chen et al.

    Cancer burden from arsenic in drinking water in Bangladesh

    Am. J. Public Health

    (2004)
  • C.J. Chen et al.

    Epidemiological characteristics and risk factors of hepatocellular carcinoma

    J. Gastroenterol. Hepatol.

    (1997)
  • H. Chen et al.

    Chronic inorganic arsenic exposure induces hepatic global and individual gene hypomethylation: implications for arsenic hepatocarcinogenesis

    Carcinogenesis

    (2004)
  • J.D. Cook et al.

    Interaction between genetic susceptibility and early life environmental exposure determines tumor suppressor gene penetrance

    Proc. Natl. Acad. Sci. U.S.A.

    (2005)
  • D. Dai et al.

    Cloning, sequencing, heterologous expression, and characterization of murine cytochrome P450 3a25*(Cyp3a25), a testosterone 6beta-hydroxylase

    J. Biochem. Mol. Toxicol.

    (2001)
  • J. Fishman et al.

    The role of estrogen in mammary carcinogenesis

    Ann. N. Y. Acad. Sci.

    (1995)
  • T. Goto et al.

    Regulation of X-chromosome inactivation in development in mice and humans

    Microbiol. Mol. Biol. Rev.

    (1998)
  • Cited by (44)

    • Environmentally relevant arsenic exposure affects morphological and molecular endpoints associated with reproduction in the Western mosquitofish, Gambusia affinis

      2022, Science of the Total Environment
      Citation Excerpt :

      Other estrogen-related outcomes following exposure to As include decreases in estradiol and related hormones (Chattopadhyay et al., 2003; Chatterjee and Chatterji, 2010), and inhibition of ER protein expression associated with diminished mass ovarian and uterine tissues (Chattopadhyay et al., 2003). Conversely, over-expression of ERα and or ERβ (Liu et al., 2007; Waalkes et al., 2004; Che et al., 2019), and upregulation of ER related proteins (Waalkes et al., 2000; Che et al., 2019) have also been observed in male tissues under some exposure conditions. These studies together with ours suggest that As may act as an endocrine disruptor influencing reproductive endpoints critical to fitness outcomes.

    • Intrauterine exposure of mice to arsenite induces abnormal and transgenerational glycometabolism

      2022, Chemosphere
      Citation Excerpt :

      In contrast, for males, changes in the F2 generation induced by exposure of F0 animals are designated transgenerational effects, because, in this case, only F0 and its germline (F1) are directly exposed (Nilsson et al., 2018; Skvortsova et al., 2018; Brehm and Flaws, 2019). Arsenic exposure can disturb the hormone systems and affect DNA methylation in offspring (Liu et al., 2007; Shen et al., 2007), demonstrating that it causes transgenerational effects. For the nematode, Caenorhabditis elegans, transgenerational reproductive effects caused by arsenic exposure have been documented up to the F3 generation (Yu and Liao, 2016).

    • Mixture effect of arsenic and fluoride at environmentally relevant concentrations in zebrafish (Danio rerio) liver: Expression pattern of Nrf2 and related xenobiotic metabolizing enzymes

      2019, Aquatic Toxicology
      Citation Excerpt :

      Arora and Bhateja (2014) observed a significant relation between mean F concentration of the soil and mean F concentration of rice and wheat. Extensive research have been conducted in recent years on the genotoxic, hepatotoxic and neurotoxic potential of As or F exposure alone in various organisms particularly in mammals and fish (Liu et al., 2007; Sarkar et al., 2017; Chattopadhyay et al., 2011; Mukhopadhyay et al., 2015; Podder et al., 2010) whereas reports on the biological response to their combined effects are rare and ambiguous. Some reports suggest that simultaneous exposure to As and F tend to neutralize each other exhibiting antagonism (Flora et al., 2009) though reports on their synergistic effects are also available (Tiwari and Rao, 2010; Jhala et al., 2008).

    View all citing articles on Scopus
    View full text