3,367
Views
178
CrossRef citations to date
0
Altmetric
Review

Environmental epigenetics in metal exposure

&
Pages 820-827 | Received 29 Apr 2011, Accepted 01 May 2011, Published online: 01 Jul 2011

Abstract

Although it is widely accepted that chronic exposure to arsenite, nickel, chromium and cadmium increases cancer incidence in individuals, the molecular mechanisms underlying their ability to transform cells remain largely unknown. Carcinogenic metals are typically weak mutagens, suggesting that genetic-based mechanisms may not be primarily responsible for metal-induced carcinogenesis. Growing evidence shows that environmental metal exposure involves changes in epigenetic marks, which may lead to a possible link between heritable changes in gene expression and disease susceptibility and development. Here, we review recent advances in the understanding of metal exposure affecting epigenetic marks and discuss establishment of heritable gene expression in metal-induced carcinogenesis.

Introduction

Chronic environmental exposure to some metal compounds, including arsenic, nickel, chromium and cadmium, has been known to induce cancers and other diseases in exposed individuals.Citation1Citation4 This ability has been long confirmed in cell culture and animal models.Citation5,Citation6 However, most of these metals, with the exception of chromium, are mildly mutagenic or non-mutagenic in conventional bacterial and mammalian mutagenesis assays.Citation7,Citation8 Their ubiquitous and indestructible nature makes these metals the most resilient form of environmental pollutants. While it is thought that these metals disturb a vast array of cellular processes, such as redox state and various intracellular stress-signaling pathways, their ability to induce diseases remains poorly understood.Citation9Citation11 Sources of potential environmental exposure to these metals include occupational exposure and environmental contamination from massive industrial production with poor practices in the disposal (i.e., the largest toxic sites known as Superfund sites in the US).Citation12

Recent evidence from animal studies shows the important role of epigenetics in response to environmental factors including nutritional supplements and xenobiotic chemicals.Citation13Citation15 Furthermore, emerging epidemiological studies show that the carcinogenic potential of some toxic metals may involve epigenetic changes, including silencing of DNA repair and tumor-suppressor genes.Citation16Citation18 It is now becoming apparent that a full understanding of the effects of environmental metals on carcinogenesis will require the consideration of epigenetic-based mechanisms in addition to genetic-based mechanisms.Citation19,Citation20

Various epigenetic marks regulate the dynamic nature of chromatin structure and are essential components for regulating heritable gene expression.Citation21,Citation22 In particular, DNA methylation and histone post-translational modifications have received much attention as mediators of epigenetic processes. Other components of chromatin are also potential mediators of epigenetic inheritance, including histone variants, non-coding RNAs, non-histone chromatin binding proteins, and the higher-order structure of chromatin itself.Citation23,Citation24 In this Review, we focus in recent evidence from experimental and epidemiological studies implicating that some toxic metal exposures can alter epigenetic marks and gene expression patterns.

Arsenic

Arsenic (As) is a widely spread environmental contaminant that can be found in the soil and water and as airborne particles. Epidemiological studies have linked arsenic exposure to the development of lung, bladder, kidney and liver cancer.Citation25,Citation26 Exposure to arsenic occurs in the form of either arsenite (AsIII) or arsenate (AsV).Citation27 The increased cancer risk to arsenic is attributed to arsenite rather than arsenate, possibly due to the cell's ability to take up arsenite at a faster rate than arsenate.Citation27,Citation28 Although arsenic has been proposed to disturb a variety of cellular processes including cellular redox status and signal transduction, recent evidence suggests that arsenic may promote cellular transformation through chromatin-based mechanisms.Citation29

DNA methylation.

Epidemiological studies show that arsenic exposure leads to gene-specific DNA hypermethylation in human subjects. In West Bengal, analysis of whole blood samples from individuals chronically exposed to arsenite in the drinking water shows that DNA hypermethylation in the promoter regions of the Cyclin-Dependent Kinase Inhibitor 2A (CDKN2A/p16INK4a) and p53 genes correlates with their gene silencingCitation30 (). Similarly, a study on a population of New Hampshire individuals with bladder cancer identifies a correlation between arsenic exposure and increased DNA methylation in the promoter regions of Ras Association Domain Family Protein 1A (RASSF1A) and Serine Protease 3 (PRSS3),Citation31 although the expression levels of these genes are not known. A recent analysis of bladder cancer samples of Taiwanese individuals living in areas with known arsenic contamination indicates a correlation between arsenic exposure and Death-Associated Protein Kinase (DAPK) promoter DNA hypermethylation with loss of DAPK expression.Citation32 Also, DNA methylation has been implicated in silencing of the p21/CDKN1A and Metallothionein-1 (MT-1) genes in cells chronically exposed to arsenite, as treatment of these cells with the DNA methyltransferase (DNMT) inhibitor 5-azacytidine (5-aza) partially restores expression of these genes.Citation33 Currently, the mechanisms whereby arsenic treatment induces gene-specific DNA hypermethylation are not well understood.

Effects on gene-specific DNA methylation have also been reported in mice chronically exposed to arsenite. Similar to the human study, silencing of the CDKN2A and RASSF1A genes is correlated with the promoter DNA hypermethylation in lung tissue and tumors from mice chronically exposed to arsenate.Citation34 In contrast to these findings, the promoters of Estrogen Receptor alpha (ER-α) and Cyclin D1 are hypomethylated in liver tissue from arsenite-exposed mice, and these genes are expressed at 3- to 5-fold higher level than control animals.Citation35 Similar DNA hypomethylation of these genes has also been reported in mice exposed to arsenic during gestation.Citation36 Interestingly, the ER-α and Cyclin D1 expression profile in men chronically exposed to arsenic is similar to that reported in mice, although the promoter DNA methylation status is not known in human samples.Citation36

Both short and chronic exposures to arsenite lead to changes in global DNA methylation.Citation37Citation39 Analysis of liver tissue from mice exposed to arsenite during gestation reveals decreased levels of DNA methylation at GC-rich areas of the genome and global deregulation of gene expression, including downregulation of members of the cytochrome p450 family and upregulation of members of the glutathione S-transferase family.Citation40 However, the mechanisms by which global DNA hypomethylation increases and decreases gene expression are not known. Also, global DNA hypomethylation by chronic exposure is correlated to the cell transformation and the ability of transformed cells to induce tumors in nude mice.Citation38 In contrast, analysis from peripheral blood of individuals chronically exposed to arsenic in the drinking water indicates mild global DNA hypermethylation.Citation41 Therefore, DNA methylation patterns by arsenite exposure are not always consistent, and the discrepancy on global DNA methylation among studies is not well understood. Arsenic detoxification generally involves methylation by the arsenic methyl transferase AS3MT that utilizes S-adenosyl methionine (SAM) as the methyl donor.Citation42 This is of importance given that SAM is the universal methyl donor for all methylation processes in the cell including DNA and histone methylation.Citation43 Therefore, high concentration of arsenite exposure can lead to reduction of global DNA methylation. This notion is generally consistent with studies in cell and animal model systems, where the concentration tends to be high. However, evidence supporting this notion is generally lacking, as the level of SAM is not concomitantly determined with DNA methylation. Moreover, most epidemiological human studies show DNA hypermethylation in a gene-specific manner, where the concentration of arsenite would be low with chronic exposure. Therefore, it is possible that the effect of arsenite on DNA methylation could be dependent on dose or other factors (i.e., AS3MT activity), which may affect the intracellular SAM levels perhaps in a cell/tissue-specific manner. Furthermore, at least in some cases, the observed global DNA hypomethylation is suggested to be a result of reduced expression of the DNMT1 and DNMT3A/B although the mechanisms behind these observations remain unclear.Citation39

Histone modifications.

Recent evidence indicates that arsenite treatment regulates gene expression through changes in histone modifications. In cells carrying the Mouse Mammary Tumor Virus (MMTV), arsenite exposure inhibits dexamethasone-induced dimethylation of arginine 17 of histone H3 (H3R17me2; a marker of transcription activation) at the nucleosome B of the MMTV promoter and suppresses its expressionCitation44 (). This is correlated with reduced SacI accessibility to the nucleosome B of the MMTV promoter as well as with inhibition of the Co-activator-Associated Arginine Methyltransferase-1 (CARM1) recruitment to this nucleosome. In addition, arsenite treatment is shown to increase the level of phosphoacetylation of histone H3 at lysine 9/serine 10 (H3K9AcS10P; a marker of transcription activation) at the promoters of the Heat Shock Protein 70 (HSP70), Mitogen-Activated Kinase Phosphatase 1 (MKP-1), c-Jun and c-Fos genes. The level of H3K9AcS10P is correlated to increased gene expression in the p38MAPK-dependent pathway.Citation11,Citation45,Citation46

Interestingly, analysis of parental and arsenite-transformed cell lines indicates upregulation of histone H3 trimethylation at lysines 4, 9 and 27 (H3K4me3, H3K9me3 and H3K27me3) and constitutive binding of the transcription factor MTF-1 at the Metallothionein-3 (MT-3) promoter.Citation47 The simultaneous presence of epigenetic marks for transcription activation (H3K4me3) and inactivation (H3K9me3 and H3K27me3) at promoters is thought to “mark” genes that are silent but poised for rapid activation.Citation48 MT-3 expression is silenced in both cell lines but can be induced by treatment with a histone deacetylase (HDAC) inhibitor. Interestingly, arsenite-transformed cells produce up to five-fold more MT-3 transcript as compared to the parental cells by treatment with the HDAC inhibitor, and this is correlated to downregulation of both H3K9me3 and H3K27me3, with concomitant upregulation of H4 acetylation and H3K4me3.Citation47 These findings indicate that arsenic-induced cell transformation may involve establishing a poised promoter chromatin state of the MT-3 gene for rapid activation and expression through heritable alterations of the promoter histone modifications.

Brief exposures to arsenite are shown to increase the global levels of histone H3 phosphorylation at serine 10 (H3S10P), acetylation at lysine 9 (H3K9Ac) and H3K9AcS10P in several systems, whereas chronic exposure increases the global levels of H3K4me3 and H3K9me2.Citation11,Citation46,Citation49Citation52 The short treatment in some of these studies suggests that the differences observed are likely to be a consequence of activated signal transduction pathways leading to the expression of the immediate stress response genes, including HSP70 and c-jun/c-fos.

Nickel

Nickel (Ni) is a toxic and carcinogenic metal of environmental concern due to its wide use in industry including the production of jewelry, coins, plating, welding, batteries and medical devices. In addition, nickel is currently used as a catalyst in carbon nanoparticle synthesis. Inhalation of nickel particles represents the major route of human exposure.Citation53 Nickel has been known to cause multiple respiratory system conditions from mild irritation and inflammation to pulmonary edema and cancers.Citation54 Due to the poor mutagenic ability of nickel compounds in bacterial and mammalian cell culture systems, it is thought that this metal exerts its toxic effects through non-genotoxic mechanisms.Citation55

DNA methylation.

The absence of chromosomal aberrations in nickel-transformed cells suggests that DNA methylation may be a mechanism of transformation by nickel treatment.Citation5,Citation56,Citation57 Furthermore, chromosome transfer experiments demonstrate that the unlimited proliferative character of a nickel-transformed cell line could be reversed by induction of senescence. The senescence gene(s) is located in the donor X-chromosome, and its expression is regulated by DNA methylation, as treatment of cells with 5-aza restores the senescence capacity of donor cells. These data indicate that transformation by nickel may involve DNA methylation and silencing of a senescence gene(s) in the X chromosome.Citation58

Recent evidence shows that nickel transformation results in silencing of the DNA repair gene O6-methylguanine DNA methyltransferase (MGMT) expression in lung cancer cells.Citation17 MGMT silencing correlates to DNA hypermethylation of its promoter (along with histone H3 deacetylation and enrichment of H3K9me2). Subsequent binding of the methyl-CpG-binding protein-2 (MeCP2) protein at the promoter indicates chromatin condensation of the MGMT promoter. Importantly, the epigenetic character of these alterations is strengthened by the fact that MGMT expression and partial reversal of repressive chromatin modifications could be achieved by treatment of cells with 5-aza and siRNA-mediated knockdown of DNMT1.Citation17 In addition, DNA hypermethylation is shown to occur at the bacterial xanthine guanine phosphoribosyltransferase (gpt) transgene promoter.Citation59 The gpt transgene is integrated near a heterochromatic area in cells but is normally expressed with gene activation-associated acetylated histones H3 and H4 in its promoter. However, transformation of these cells with nickel results in gpt silencing associated with increased DNA methylation and chromatin condensation.Citation59 In addition, nickel treatment leads to subsequent binding of MeCP2 at the gpt promoter.Citation60,Citation61 Importantly, these phenomena could be partially reverted by pretreatment of cells with 5-aza, further implicating that nickel treatment may exert gene silencing through altering DNA methylation.Citation59,Citation60

A small number of studies have focused on the direct effects of nickel on mononucleosome and nucleosome array structures. X-ray crystal structures of reconstituted mononucleosomes in the presence of millimolar concentrations of nickel reveal up to 43 putative metal-binding sites in the nucleosome with preference towards GG motifs.Citation62 Nickel binding induces alterations in the DNA-histone interaction through stretching of the DNA near the dyad axis, resulting in protrusion of two DNA bases off the nucleosome. These alterations are thought to condense higher-order chromatin structures. Indeed, previous studies using micrococcal nuclease-digested chromatin incubated with various divalent cations indicate that nickel promotes nucleosome array condensation at lower concentrations than magnesium.Citation63 Additional evidence from atomic force microscopy and circular dichroism measurements suggests that nickel is effective at promoting condensation of reconstituted nucleosome arrays.Citation64

Histone modifications.

Evidence indicates that the Sprouty Homolog 2 (Spry2), gpt and MGMT genes are linked to nickel-induced gene silencing with changes in histone modifications.Citation17,Citation59,Citation65 Nickel treatment inhibits expression of the Spry2 gene, a negative regulator of receptor tyrosine kinases.Citation66 The Spry2 silencing is correlated with increased level of H3K9me2, a marker of transcription inactivation, at its promoter.Citation65 Interestingly, increased level of H3K9me2 at the Spry2 promoter is thought to arise because of nickel inhibition of the H3K9-specific, iron-dependent histone demethylase Jumonji domain-containing protein 1A (JMJD1A). In vitro, nickel directly inactivates JMJD1A at an equal molar ratio, suggesting a mechanism involving iron replacement in the catalytic center.Citation67

Similar to results from the DNA methylation study of the gpt transgene, transformation of these cells with nickel results in gpt silencing associated with histone deacetylation, increased level of H3K9me2 and chromatin condensation.Citation59 These events can be partially reversed by co-treatment of cells with the HDAC inhibitor Trichostatin A (TSA), implicating that nickel-induced gpt silencing involves modulation of the local gpt chromatin structure. This notion is reinforced by the fact that gpt is insensitive to nickel-induced silencing if integrated away from heterochromatic areas.Citation68 However, alterations of epigenetic marks at the gpt promoter chromatin triggered by nickel exposure may only be temporal, as removal of nickel from media eventually leads to expression of gpt.Citation69 Also, similar to results from the DNA methylation study, the MGMT gene silencing correlates to histone H3 deacetylation and increased H3K9me2, which is partially reversed by treatment of cells with TSA.Citation17

Nickel exposure has also been shown to affect the global levels of histone modifications, implicating global deregulation of gene expression. Treatment of nickel-transformed cells with TSA has been shown to reverse the phenotype to that of untransformed cells along with the gene expression profile, suggesting that histone acetylation might be inhibited during nickel-induced transformation.Citation70 However, it is not clear whether this reversible transforming phenotype is associated with the altered gene expression profile. Further support for nickel inhibition of histone acetylation comes from experiments where continuous nickel exposure leads to histone H4 hypoacetylation in yeast and human cells.Citation71 Nickel binding to the H4 N-terminal peptide at His 18 is thought to interfere with the interaction of histone acetyltransferase complexes (HAT) with chromatin.Citation72

Other histone modifications that are also globally affected by nickel treatment include H3K9me2, H3K4me3 and ubiquitination of histones H2A and H2B (H2A/H2Bub).Citation49,Citation60,Citation61,Citation73,Citation74 In some cases, the increase in H2A/H2B ubiquitination has been correlated to upregulation of the Ubiquitin-Conjugating Enzyme H6 (UbcH6) E2 ligase at the protein level or inhibition of an unidentified histone deubiquitinating activity.Citation73,Citation74 However, the mechanisms whereby nickel affects the levels of these global histone modifications and their significance on epigenetic heritance remain unexplored.

Extended exposure to nickel is shown to result in cleavage of C- and N-terminal peptides of H2B and a C-terminal octapeptide of H2A, possibly due to nickel-induced histone oxidation.Citation75Citation77 Interestingly, nickel has been shown to form a stable complex with the C-terminal ELAKHA hexapeptide of H2B.Citation78 Currently, the mechanism of peptide cleavage and its biological significance remain elusive.

Chromium

Chromium (Cr) exposure to humans is widespread, given that multiple industries including stainless steel welding, chrome plating and ferrochrome manufacturing utilize this metal. Chromium is also found in the environment in the form of airborne particles arising from automobile catalytic converters.Citation12 Chromium exposure has been linked to respiratory conditions including nasal ulceration and lung cancers.Citation2,Citation79 The major source of exposure for humans is chromium-contaminated water, typically located in the vicinity of industrial areas. It is speculated that tens of millions of people are exposed to chromium contamination worldwide. Although chromium can be found in various oxidative states including +6, +3 and 0, Cr(VI) is the prevalent form of chromium that has been epidemiologically linked to development of respiratory cancers.Citation12 The major mechanisms of chromium-induced cytotoxicity are thought to be genotoxic, as it is well documented that chromium can lead to oxidative stress, DNA strand breaks, DNA-protein crosslinks and formation of stable chromium-DNA adducts.Citation80,Citation81

DNA methylation.

In lung cancer specimens of individuals chronically exposed to chromium, silencing of the mismatch repair gene Human MutL Homolog 1 (hMLH1) and the tumor suppressor p16INK4a is correlated with DNA hypermethylation of their promoters.Citation18,Citation82 Furthermore, DNA hypermethylation in the promoter regions of the Adenomatous Polyposis Coli (APC) tumor suppressor and MGMT is detected in these samples, although a correlation with their gene expression levels is not established.Citation16 Chromium has also been found to induce DNA hypermethylation of the gpt promoter and subsequent silencing of its expression in cells.Citation83

Histone modifications.

Similar to results of the DNA methylation study from human samples, chromium exposure results in increased level of H3K9me2 at the promoter of hMLH1 gene and reduces its expression in lung cancer cells.Citation84 Interestingly, the increased level of H3K9me2 is correlated to upregulation of the H3K9 methylase G9a at both mRNA and protein levels in these cells.Citation84 In addition, short exposure to chromium leads to a reduction in the levels of H3K4me3 and H3/H4 Ac and blocks aryl hydrocarbon receptor (AhR) and RNAP II recruitment at the Cytochrome p450 Subfamily 1 Polypeptide 1 (CYP1a1) promoter, with the corresponding inhibition of CYP1a1 expression in response to benzo(α)pyrene.Citation85 These events are shown to correlate to the chromium-dependent crosslinking of DNMT1 and HDAC1 at the CYP1a1 promoter, suggesting that reduction of H3K4me3 and H3/H4 Ac levels at the CYP1a1 promoter is likely a result from the physical obstruction of repressor complexes. This in turn may prevent access of histone-modifying complexes and the transcriptional machinery, thus resulting in reduced CYP1a1 expression. In addition, short chromium exposure increases the global levels of H3K4me3 and H3K9me2.Citation49 Both modifications appear to be differentially localized in the chromatin regions and have been speculated to alter gene expression patterns.

Cadmium

Cadmium (Cd) is a toxic transition metal that is both an environmental and occupational toxicant. Sources of cadmium exposure include battery manufacturing, some electroplating processes and consumption of tobacco products. Cadmium has been designated a human carcinogen by both the International Agency for Cancer Research and the National Toxicology program.Citation86 Multiple epidemiological studies have linked cadmium exposure with pulmonary cancers, while fewer studies have linked it to bladder and pancreatic cancers.Citation87 In addition, cadmium is a multiple tissue carcinogen in animal models.Citation88 Its long half-life and poor excretion make cadmium a cumulative toxin. As such, previous exposures to cadmium may lead to detrimental effects during the lifetime of exposed individuals. Although the mechanisms of cadmium-induced transformation are not understood, its effects are thought to arise through the disruption of zinc-dependent cellular processes.Citation89 This is due to the structural and physical similarities between zinc and cadmium. In addition, although cadmium can produce oxidative stress, it is poorly mutagenic and has a weak DNA binding affinity. This has forwarded the hypothesis that cadmium may promote carcinogenesis through epigenetic mechanisms.Citation86

DNA methylation.

Gene-specific DNA hypermethylation and gene silencing have been observed in cells chronically exposed to cadmium, involving the p16INK4a, RASSF1A and MT-1 genes.Citation90,Citation91 Importantly, expression of these genes can be restored by treatment of cells with 5-aza, further strengthening the correlation between DNA methylation and gene silencing. Increased expression of DNMT3B is shown to correlate with DNA hypermethylation of the p16INK4a and RASFF1A promoters.Citation90 However, details on the mechanisms of MT-1 silencing are lacking, as this event is only correlated to increased total cellular DNMT activity.Citation91 In addition, recent evidence indicates that global DNA methylation is affected by cadmium exposure. Short exposures to cadmium lead to reduction of global de novo DNA methylation in chick embryos.Citation92 The reduced DNA methylation levels are thought to be a result of the inhibitory effects of cadmium on expression of DNMT3A/B, the enzymes responsible for de novo DNA methylation during chick development. Another report suggests that DNA hypomethylation resulting from short exposures to cadmium may potentially be correlated with increased cell cycle rates in cells, although the evidence of a direct connection between these two events is lacking.Citation93 Short exposures to cadmium is to a dose-dependent reduction on DNMT activity, resulting in DNA hypomethylation. In contrast, chronic exposures result in a dose-dependent increase in cellular DNMT activity and concomitant DNA hypermethylation, even after cadmium is removed from the medium.Citation91 However, the discrepancy on global DNA methylation patterns between short and long exposures to cadmium in this study is not well understood. In addition, global DNA hypermethylation has been correlated with upregulation of DNMT1 and DNMT3A/B expression in cells transformed by and chronically exposed to cadmium.Citation90,Citation94 The overall pattern suggests that short exposures are associated with DNA hypomethylation, whereas chronic exposure is associated with DNA hypermethylation.

Histone modifications.

Evidence suggests that cadmium exposure may lead to heritable changes in chromatin structure for rapid transcription activation. Transformation of cells by cadmium results in the establishment and maintenance of a bivalent chromatin domain at the MT-3 promoter, involving changes in histone modifications nearly identical to those observed for arsenic-transformed cells.Citation47 This inheritable alteration of chromatin structure leads to inducibility of MT-3 expression when cells are treated with an HDAC inhibitor. Therefore, these data indicate that transformation by cadmium may establish and stably maintain chromatin structure of the MT-3 promoter for rapid gene activation.Citation47

Mercury

Although strictly not considered a metal, mercury is an environmental toxicant that is correlated with brain toxicity. Indeed, it has been long known that exposure to mercury results in defects in brain function.Citation95 Due to its ability to cross the placenta, mercury is thought to be most toxic during the development of the fetus.Citation96 In cell culture models, methylmercury (MeHg) affects expression of several neuronal growth factors, therefore affecting neural stem cell differentiation and neurite outgrowth.Citation97Citation99 Various types of seafood contain high levels of mercury and become a source of exposure.

Histone modifications.

Prenatal methylmercury exposure increases the level of H3K27me3 and decreases the level of H3Ac at the Brain-Derived Neurotrophic Factor (BDNF) promoter, which results in reduced expression of BDNF in the dentate gyrus.Citation100 This is correlated to a depression-like behavioral phenotype. Interestingly, treatment with the anti-depressant fluorexine restores BDNF expression in the dentate gyrus, and this correlates to upregulation of H3Ac, without affecting H3K27me3, at the BDNF promoter.Citation100 Restoration of BDNF expression also correlates to reversal of the depression-like behavior. These results imply that a methylmercury-induced epigenetic silencing of the BDNF in the hippocampus may contribute to a behavioral phenotype.

Conclusion and Future Perspectives

Accumulating evidence clearly shows that toxic metal exposures lead to the induction and alteration of epigenetic marks in experimental and epidemiological studies. These studies highlight the possibility that environmental metals may exert their effects on gene expression through establishing and maintaining epigenetic chromatin states. Currently, it is not clear whether these altered epigenetic marks are linked to heritable gene expression patterns or the causative pathway to the disease (cancer) susceptibility and development.

To establish that epigenetic mechanisms underlie the molecular mechanisms of actions for metal exposures, there are a number of factors to be investigated in the future. Changes in epigenetic marks at the global level require further evaluation for their involvement in epigenetic processes. It is important to eliminate the possibility that alterations in global DNA methylation, or any other global epigenetic marks, are not a reflection of dynamic global cellular responses. This is especially critical for metal exposure, where metals can affect multiple cellular processes, including global chromatin structure. For instance, nickel is proposed to bind to and condense chromatin, and chromium can crosslink chromatin-associated proteins to DNA.Citation62Citation64,Citation85,Citation101,Citation102 Also, cadmium is shown to potentially promote global DNA hypermethylation by either stimulating DNMT activity or inducing its gene expression, whereas arsenite can deplete SAM, leading to DNA hypomethylation.Citation37Citation39,Citation42,Citation90 Furthermore, studies of arsenite show that global DNA hypomethylation does not always correlate with local gene-specific DNA hypermethylation or gene expression patterns, suggesting that a gene-by-gene-based analysis is required for establishing epigenetic-based mechanisms.Citation29,Citation30 Given that multiple cellular processes are affected by exposure to these metals, identification of the initial epigenetic signaling pathways or factors that establish and maintain metal-induced heritable chromatin states could provide further evidence for epigenetic mechanisms.

Most of the global and local epigenetic marks mediated by metal exposure in experimental studies are generally measurable only in the continuous presence of metals. Removal of nickel treatment leads to re-expression of a nickel-silenced gene, therefore suggesting that altered epigenetic marks could be a result of dynamic cellular responses.Citation69 Also, alterations in epigenetic marks induced by brief or short-term exposure most likely represent ongoing signal transduction and may not represent heritable epigenetic events, especially without evidence of heritable epigenetic marks correlating with gene expression patterns. In the case of cadmium, short- and long-term exposures result in the opposite effect on DNA methylation through different mechanisms, suggesting that duration, in addition to dose, of exposure affects the outcome of epigenetic marks.Citation69,Citation92Citation94 The only exception to this is the study of the MT-3 gene.Citation47 This study suggests that regulation of epigenetic events by transient exposures to arsenite and cadmium may not necessarily involve heritable gene expression patterns but the establishment and maintenance of heritable epigenetic chromatin states, which poise the gene for rapid activation in response to a stimulus.

Currently, it is not clear whether specific gene silencing and associated with epigenetic marks induced by metals are transitory or heritable in both experimental and epidemiological studies. Although studies are limited, it would be of great importance to determine whether silencing of tumor suppressor and DNA repair genes could be identified as a common set of epigenetically labile genes that could play roles in metal-induced carcinogenesis. Furthermore, this identification would provide epigenetic biomarkers for detecting early-stages of cancer.

Collectively, an increasing body of evidence clearly supports the concept that epigenetics has potential for better understanding of the molecular mechanisms whereby environmental metal exposure leads to heritable epigenetic marks, correlating with gene expression patterns. Future studies of epigenetic marks need to address the heritability of these marks and correlate them to heritable gene expression patterns, possibly linking them to metal-induced carcinogenesis.

Figures and Tables

Table 1 Metal-induced epigenetic marks

Acknowledgements

This work was supported by funding from the National Institutes of Health (grants ES015210 and AI64706) to H.C.H.

References

  • Doll R, Morgan LG, Speizer FE. Cancers of the lung and nasal sinuses in nickel workers. Br J Cancer 1970; 24:623 - 632
  • Gibb HJ, Lees PS, Pinsky PF, Rooney BC. Lung cancer among workers in chromium chemical production. Am J Ind Med 2000; 38:115 - 126
  • Smith AH, Hopenhayn-Rich C, Bates MN, Goeden HM, Hertz-Picciotto I, Duggan HM, et al. Cancer risks from arsenic in drinking water. Environ Health Perspect 1992; 97:259 - 267
  • Yuan Y, Marshall G, Ferreccio C, Steinmaus C, Selvin S, Liaw J, et al. Acute myocardial infarction mortality in comparison with lung and bladder cancer mortality in arsenic-exposed region II of Chile from 1950 to 2000. Am J Epidemiol 2007; 166:1381 - 1391
  • Biedermann KA, Landolph JR. Induction of anchorage independence in human diploid foreskin fibroblasts by carcinogenic metal salts. Cancer Res 1987; 47:3815 - 3823
  • Dunnick JK, Benson JM, Hobbs CH, Hahn FF, Cheng YS, Eidson AF. Comparative toxicity of nickel oxide, nickel sulfate hexahydrate and nickel subsulfide after 12 days of inhalation exposure to F344/N rats and B6C3F1 mice. Toxicology 1988; 50:145 - 156
  • Biggart NW, Costa M. Assessment of the uptake and mutagenicity of nickel chloride in salmonella tester strains. Mutat Res 1986; 175:209 - 215
  • Kerckaert GA, LeBoeuf RA, Isfort RJ. Use of the Syrian hamster embryo cell transformation assay for determining the carcinogenic potential of heavy metal compounds. Fundam Appl Toxicol 1996; 34:67 - 72
  • Andrew AS, Klei LR, Barchowsky A. Nickel requires hypoxia-inducible factor-1 alpha, not redox signaling, to induce plasminogen activator inhibitor-1. Am J Physiol Lung Cell Mol Physiol 2001; 281:607 - 615
  • Fowler BA, Whittaker MH, Lipsky M, Wang G, Chen XQ. Oxidative stress induced by lead, cadmium and arsenic mixtures: 30-day, 90-day and 180-day drinking water studies in rats: an overview. Biometals 2004; 17:567 - 568
  • Thomson S, Hollis A, Hazzalin CA, Mahadevan LC. Distinct stimulus-specific histone modifications at hsp70 chromatin targeted by the transcription factor heat shock factor-1. Mol Cell 2004; 15:585 - 594
  • Salnikow K, Zhitkovich A. Genetic and epigenetic mechanisms in metal carcinogenesis and cocarcinogenesis: nickel, arsenic and chromium. Chem Res Toxicol 2008; 21:28 - 44
  • Anway MD, Cupp AS, Uzumcu M, Skinner MK. Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 2005; 308:1466 - 1469
  • Cropley JE, Suter CM, Beckman KB, Martin DI. Germ-line epigenetic modification of the murine A vy allele by nutritional supplementation. Proc Natl Acad Sci USA 2006; 103:17308 - 17312
  • Dolinoy DC, Huang D, Jirtle RL. Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development. Proc Natl Acad Sci USA 2007; 104:13056 - 13061
  • Ali AH, Kondo K, Namura T, Senba Y, Takizawa H, Nakagawa Y, et al. Aberrant DNA methylation of some tumor suppressor genes in lung cancers from workers with chromate exposure. Mol Carcinog 2011; 50:89 - 99
  • Ji W, Yang L, Yu L, Yuan J, Hu D, Zhang W, et al. Epigenetic silencing of O6-methylguanine DNA methyltransferase gene in NiS-transformed cells. Carcinogenesis 2008; 29:1267 - 1275
  • Kondo K, Takahashi Y, Hirose Y, Nagao T, Tsuyuguchi M, Hashimoto M, et al. The reduced expression and aberrant methylation of p16(INK4a) in chromate workers with lung cancer. Lung Cancer 2006; 53:295 - 302
  • Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet 2007; 8:253 - 262
  • Szyf M. The dynamic epigenome and its implications in toxicology. Toxicol Sci 2007; 100:7 - 23
  • Kouzarides T. Chromatin modifications and their function. Cell 2007; 128:693 - 705
  • Margueron R, Reinberg D. Chromatin structure and the inheritance of epigenetic information. Nat Rev Genet 2010; 11:285 - 296
  • Probst AV, Dunleavy E, Almouzni G. Epigenetic inheritance during the cell cycle. Nat Rev Mol Cell Biol 2009; 10:192 - 206
  • Talbert PB, Henikoff S. Spreading of silent chromatin: inaction at a distance. Nat Rev Genet 2006; 7:793 - 803
  • Hopenhayn-Rich C, Biggs ML, Fuchs A, Bergoglio R, Tello EE, Nicolli H, et al. Bladder cancer mortality associated with arsenic in drinking water in Argentina. Epidemiology 1996; 7:117 - 124
  • Marshall G, Ferreccio C, Yuan Y, Bates MN, Steinmaus C, Selvin S, et al. Fifty-year study of lung and bladder cancer mortality in Chile related to arsenic in drinking water. J Natl Cancer Inst 2007; 99:920 - 928
  • Bertolero F, Pozzi G, Sabbioni E, Saffiotti U. Cellular uptake and metabolic reduction of pentavalent to trivalent arsenic as determinants of cytotoxicity and morphological transformation. Carcinogenesis 1987; 8:803 - 808
  • Lerman SA, Clarkson TW, Gerson RJ. Arsenic uptake and metabolism by liver cells is dependent on arsenic oxidation state. Chem Biol Interact 1983; 45:401 - 406
  • Reichard JF, Puga A. Effects of arsenic exposure on DNA methylation and epigenetic gene regulation. Epigenomics 2010; 2:87 - 104
  • Chanda S, Dasgupta UB, Guhamazumder D, Gupta M, Chaudhuri U, Lahiri S, et al. DNA hypermethylation of promoter of gene p53 and p16 in arsenic-exposed people with and without malignancy. Toxicol Sci 2006; 89:431 - 437
  • Marsit CJ, Karagas MR, Danaee H, Liu M, Andrew A, Schned A, et al. Carcinogen exposure and gene promoter hypermethylation in bladder cancer. Carcinogenesis 2006; 27:112 - 116
  • 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. Histopathology 2007; 51:785 - 792
  • Liu J, Benbrahim-Tallaa L, Qian X, Yu L, Xie Y, Boos J, et al. Further studies on aberrant gene expression associated with arsenic-induced malignant transformation in rat liver TRL1215 cells. Toxicol Appl Pharmacol 2006; 216:407 - 415
  • Cui X, Wakai T, Shirai Y, Hatakeyama K, Hirano S. Chronic oral exposure to inorganic arsenate interferes with methylation status of p16INK4a and RASSF1A and induces lung cancer in A/J mice. Toxicol Sci 2006; 91:372 - 381
  • Chen H, Li S, Liu J, Diwan BA, Barrett JC, Waalkes MP. Chronic inorganic arsenic exposure induces hepatic global and individual gene hypomethylation: implications for arsenic hepatocarcinogenesis. Carcinogenesis 2004; 25:1779 - 1786
  • Waalkes MP, Liu J, Chen H, Xie Y, Achanzar WE, Zhou YS, et al. Estrogen signaling in livers of male mice with hepatocellular carcinoma induced by exposure to arsenic in utero. J Natl Cancer Inst 2004; 96:466 - 474
  • Zhong CX, Mass MJ. Both hypomethylation and hypermethylation of DNA associated with arsenite exposure in cultures of human cells identified by methylation-sensitive arbitrarily-primed PCR. Toxicol Lett 2001; 122:223 - 234
  • Zhao CQ, Young MR, Diwan BA, Coogan TP, Waalkes MP. Association of arsenic-induced malignant transformation with DNA hypomethylation and aberrant gene expression. Proc Natl Acad Sci USA 1997; 94:10907 - 10912
  • Reichard JF, Schnekenburger M, Puga A. Long term low-dose arsenic exposure induces loss of DNA methylation. Biochem Biophys Res Commun 2007; 352:188 - 192
  • Xie Y, Liu J, Benbrahim-Tallaa L, Ward JM, Logsdon D, Diwan BA, et al. Aberrant DNA methylation and gene expression in livers of newborn mice transplacentally exposed to a hepatocarcinogenic dose of inorganic arsenic. Toxicology 2007; 236:7 - 15
  • Majumdar S, Chanda S, Ganguli B, Mazumder DN, Lahiri S, Dasgupta UB. Arsenic exposure induces genomic hypermethylation. Environ Toxicol 2010; 25:315 - 318
  • Gebel TW. Arsenic methylation is a process of detoxification through accelerated excretion. Int J Hyg Environ Health 2002; 205:505 - 508
  • Loenen WA. S-adenosylmethionine: jack of all trades and master of everything?. Biochem Soc Trans 2006; 34:330 - 333
  • Barr FD, Krohmer LJ, Hamilton JW, Sheldon LA. Disruption of histone modification and CARM1 recruitment by arsenic represses transcription at glucocorticoid receptor-regulated promoters. PLoS One 2009; 4:6766
  • Li J, Gorospe M, Barnes J, Liu Y. Tumor promoter arsenite stimulates histone H3 phosphoacetylation of proto-oncogenes c-fos and c-jun chromatin in human diploid fibroblasts. J Biol Chem 2003; 278:13183 - 13191
  • Li J, Gorospe M, Hutter D, Barnes J, Keyse SM, Liu Y. Transcriptional induction of MKP-1 in response to stress is associated with histone H3 phosphorylation-acetylation. Mol Cell Biol 2001; 21:8213 - 8224
  • Somji S, Garrett SH, Toni C, Zhou XD, Zheng Y, Ajjimaporn A, et al. Differences in the epigenetic regulation of MT-3 gene expression between parental and Cd+2 or As+3 transformed human urothelial cells. Cancer Cell Int 2011; 11:2
  • Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 2006; 125:315 - 326
  • Zhou X, Li Q, Arita A, Sun H, Costa M. Effects of nickel, chromate and arsenite on histone 3 lysine methylation. Toxicol Appl Pharmacol 2009; 236:78 - 84
  • He Z, Ma WY, Liu G, Zhang Y, Bode AM, Dong Z. Arsenite-induced phosphorylation of histone H3 at serine 10 is mediated by Akt1, extracellular signal-regulated kinase 2 and p90 ribosomal S6 kinase 2 but not mitogen- and stress-activated protein kinase 1. J Biol Chem 2003; 278:10588 - 10593
  • Kannan-Thulasiraman P, Katsoulidis E, Tallman MS, Arthur JS, Platanias LC. Activation of the mitogen- and stress-activated kinase 1 by arsenic trioxide. J Biol Chem 2006; 281:22446 - 22452
  • Ramirez T, Brocher J, Stopper H, Hock R. Sodium arsenite modulates histone acetylation, histone deacetylase activity and HMGN protein dynamics in human cells. Chromosoma 2008; 117:147 - 157
  • Kasprzak KS, Sunderman FW Jr, Salnikow K. Nickel carcinogenesis. Mutat Res 2003; 533:67 - 97
  • Morgan LG, Usher V. Health problems associated with nickel refining and use. Ann Occup Hyg 1994; 38:189 - 198
  • Fletcher GG, Rossetto FE, Turnbull JD, Nieboer E. Toxicity, uptake and mutagenicity of particulate and soluble nickel compounds. Environ Health Perspect 1994; 102:69 - 79
  • Rani AS, Qu DQ, Sidhu MK, Panagakos F, Shah V, Klein KM, et al. Transformation of immortal, non-tumorigenic osteoblast-like human osteosarcoma cells to the tumorigenic phenotype by nickel sulfate. Carcinogenesis 1993; 14:947 - 953
  • Trott DA, Cuthbert AP, Overell RW, Russo I, Newbold RF. Mechanisms involved in the immortalization of mammalian cells by ionizing radiation and chemical carcinogens. Carcinogenesis 1995; 16:193 - 204
  • Klein CB, Conway K, Wang XW, Bhamra RK, Lin XH, Cohen MD, et al. Senescence of nickel-transformed cells by an X chromosome: possible epigenetic control. Science 1991; 251:796 - 799
  • Lee YW, Klein CB, Kargacin B, Salnikow K, Kitahara J, Dowjat K, et al. Carcinogenic nickel silences gene expression by chromatin condensation and DNA methylation: a new model for epigenetic carcinogens. Mol Cell Biol 1995; 15:2547 - 2557
  • Chen H, Ke Q, Kluz T, Yan Y, Costa M. Nickel ions increase histone H3 lysine 9 dimethylation and induce transgene silencing. Mol Cell Biol 2006; 26:3728 - 3737
  • Yan Y, Kluz T, Zhang P, Chen HB, Costa M. Analysis of specific lysine histone H3 and H4 acetylation and methylation status in clones of cells with a gene silenced by nickel exposure. Toxicol Appl Pharmacol 2003; 190:272 - 277
  • Mohideen K, Muhammad R, Davey CA. Perturbations in nucleosome structure from heavy metal association. Nucleic Acids Res 2010; 38:6301 - 6311
  • Borochov N, Ausio J, Eisenberg H. Interaction and conformational changes of chromatin with divalent ions. Nucleic Acids Res 1984; 12:3089 - 3096
  • Ellen TP, Kluz T, Harder ME, Xiong J, Costa M. Heterochromatinization as a potential mechanism of nickel-induced carcinogenesis. Biochemistry 2009; 48:4626 - 4632
  • Chen H, Kluz T, Zhang R, Costa M. Hypoxia and nickel inhibit histone demethylase JMJD1A and repress Spry2 expression in human bronchial epithelial BEAS-2B cells. Carcinogenesis 2010; 31:2136 - 2144
  • Cabrita MA, Christofori G. Sprouty proteins, masterminds of receptor tyrosine kinase signaling. Angiogenesis 2008; 11:53 - 62
  • Chen H, Giri NC, Zhang R, Yamane K, Zhang Y, Maroney M, et al. Nickel ions inhibit histone demethylase JMJD1A and DNA repair enzyme ABH2 by replacing the ferrous iron in the catalytic centers. J Biol Chem 2010; 285:7374 - 7383
  • Klein CB, Costa M. DNA methylation, heterochromatin and epigenetic carcinogens. Mutat Res 1997; 386:163 - 180
  • Ke Q, Davidson T, Chen H, Kluz T, Costa M. Alterations of histone modifications and transgene silencing by nickel chloride. Carcinogenesis 2006; 27:1481 - 1488
  • Zhang Q, Salnikow K, Kluz T, Chen LC, Su WC, Costa M. Inhibition and reversal of nickel-induced transformation by the histone deacetylase inhibitor trichostatin A. Toxicol Appl Pharmacol 2003; 192:201 - 211
  • Broday L, Peng W, Kuo MH, Salnikow K, Zoroddu M, Costa M. Nickel compounds are novel inhibitors of histone H4 acetylation. Cancer Res 2000; 60:238 - 241
  • Zoroddu MA, Schinocca L, Kowalik-Jankowska T, Kozlowski H, Salnikow K, Costa M. Molecular mechanisms in nickel carcinogenesis: modeling Ni(II) binding site in histone H4. Environ Health Perspect 2002; 110:719 - 723
  • Karaczyn AA, Golebiowski F, Kasprzak KS. Ni(II) affects ubiquitination of core histones H2B and H2A. Exp Cell Res 2006; 312:3252 - 3259
  • Ke Q, Ellen TP, Costa M. Nickel compounds induce histone ubiquitination by inhibiting histone deubiquitinating enzyme activity. Toxicol Appl Pharmacol 2008; 228:190 - 199
  • Karaczyn AA, Golebiowski F, Kasprzak KS. Truncation, deamidation and oxidation of histone H2B in cells cultured with nickel(II). Chem Res Toxicol 2005; 18:1934 - 1942
  • Bal W, Liang R, Lukszo J, Lee SH, Dizdaroglu M, Kasprzak KS. Ni(II) specifically cleaves the C-terminal tail of the major variant of histone H2A and forms an oxidative damage-mediating complex with the cleaved-off octapeptide. Chem Res Toxicol 2000; 13:616 - 624
  • Karaczyn AA, Bal W, North SL, Bare RM, Hoang VM, Fisher RJ, et al. The octapeptidic end of the C-terminal tail of histone H2A is cleaved off in cells exposed to carcinogenic nickel(II). Chem Res Toxicol 2003; 16:1555 - 1559
  • Karavelas T, Mylonas M, Malandrinos G, Plakatouras JC, Hadjiliadis N, Mlynarz P, et al. Coordination properties of Cu(II) and Ni(II) ions towards the C-terminal peptide fragment-ELAKHA-of histone H2B. J Inorg Biochem 2005; 99:606 - 615
  • Gibb HJ, Lees PS, Pinsky PF, Rooney BC. Clinical findings of irritation among chromium chemical production workers. Am J Ind Med 2000; 38:127 - 131
  • Shi H, Hudson LG, Liu KJ. Oxidative stress and apoptosis in metal ion-induced carcinogenesis. Free Radic Biol Med 2004; 37:582 - 593
  • Zhitkovich A. Importance of chromium-DNA adducts in mutagenicity and toxicity of chromium(VI). Chem Res Toxicol 2005; 18:3 - 11
  • Takahashi Y, Kondo K, Hirose T, Nakagawa H, Tsuyuguchi M, Hashimoto M, et al. Microsatellite instability and protein expression of the DNA mismatch repair gene, hMLH1, of lung cancer in chromate-exposed workers. Mol Carcinog 2005; 42:150 - 158
  • Klein CB, Su L, Bowser D, Leszczynska J. Chromate-induced epimutations in mammalian cells. Environ Health Perspect 2002; 110:739 - 743
  • Sun H, Zhou X, Chen H, Li Q, Costa M. Modulation of histone methylation and MLH1 gene silencing by hexavalent chromium. Toxicol Appl Pharmacol 2009; 237:258 - 266
  • Schnekenburger M, Talaska G, Puga A. Chromium cross-links histone deacetylase 1-DNA methyltransferase 1 complexes to chromatin, inhibiting histone-remodeling marks critical for transcriptional activation. Mol Cell Biol 2007; 27:7089 - 7101
  • Arita A, Costa M. Epigenetics in metal carcinogenesis: nickel, arsenic, chromium and cadmium. Metallomics 2009; 1:222 - 228
  • Waalkes MP. Cadmium carcinogenesis in review. J Inorg Biochem 2000; 79:241 - 244
  • Waalkes MP, Anver MR, Diwan BA. Chronic toxic and carcinogenic effects of oral cadmium in the Noble (NBL/Cr) rat: induction of neoplastic and proliferative lesions of the adrenal, kidney, prostate and testes. J Toxicol Environ Health A 1999; 58:199 - 214
  • Waalkes MP. Cadmium carcinogenesis. Mutat Res 2003; 533:107 - 120
  • Benbrahim-Tallaa L, Waterland RA, Dill AL, Webber MM, Waalkes MP. Tumor suppressor gene inactivation during cadmium-induced malignant transformation of human prostate cells correlates with overexpression of de novo DNA methyltransferase. Environ Health Perspect 2007; 115:1454 - 1459
  • Takiguchi M, Achanzar WE, Qu W, Li G, Waalkes MP. Effects of cadmium on DNA-(Cytosine-5) methyltransferase activity and DNA methylation status during cadmium-induced cellular transformation. Exp Cell Res 2003; 286:355 - 365
  • Doi T, Puri P, McCann A, Bannigan J, Thompson J. Epigenetic effect of cadmium on global de novo DNA hypomethylation in the cadmium-induced ventral body wall defect (VBWD) in the chick model. Toxicol Sci 2011; 120:475 - 480
  • Huang D, Zhang Y, Qi Y, Chen C, Ji W. Global DNA hypomethylation, rather than reactive oxygen species (ROS), a potential facilitator of cadmium-stimulated K562 cell proliferation. Toxicol Lett 2008; 179:43 - 47
  • Jiang G, Xu L, Song S, Zhu C, Wu Q, Zhang L, et al. Effects of long-term low-dose cadmium exposure on genomic DNA methylation in human embryo lung fibroblast cells. Toxicology 2008; 244:49 - 55
  • Johansson C, Castoldi AF, Onishchenko N, Manzo L, Vahter M, Ceccatelli S. Neurobehavioural and molecular changes induced by methylmercury exposure during development. Neurotox Res 2007; 11:241 - 260
  • Vahter M, Akesson A, Lind B, Bjors U, Schutz A, Berglund M. Longitudinal study of methylmercury and inorganic mercury in blood and urine of pregnant and lactating women, as well as in umbilical cord blood. Environ Res 2000; 84:186 - 194
  • Parran DK, Barone S Jr, Mundy WR. Methylmercury decreases NGF-induced TrkA autophosphorylation and neurite outgrowth in PC12 cells. Brain Res Dev Brain Res 2003; 141:71 - 81
  • Tamm C, Duckworth J, Hermanson O, Ceccatelli S. High susceptibility of neural stem cells to methylmercury toxicity: effects on cell survival and neuronal differentiation. J Neurochem 2006; 97:69 - 78
  • Larkfors L, Oskarsson A, Sundberg J, Ebendal T. Methylmercury induced alterations in the nerve growth factor level in the developing brain. Brain Res Dev Brain Res 1991; 62:287 - 291
  • Onishchenko N, Karpova N, Sabri F, Castren E, Ceccatelli S. Long-lasting depression-like behavior and epigenetic changes of BDNF gene expression induced by perinatal exposure to methylmercury. J Neurochem 2008; 106:1378 - 1387
  • Costa M. DNA-protein complexes induced by chromate and other carcinogens. Environ Health Perspect 1991; 92:45 - 52
  • Misra RR, Smith GT, Waalkes MP. Evaluation of the direct genotoxic potential of cadmium in four different rodent cell lines. Toxicology 1998; 126:103 - 114

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.