Abstract
Remarkable progress in the field of epigenetics has turned academic, medical and public attention to the potential applications of these new advances in medicine and various fields of biomedical research. The result is a broader appreciation of epigenetic phenomena in the a etiology of common human diseases, most notably cancer. These advances also represent an exciting opportunity to incorporate epigenetics and epigenomics into carcinogen identification and safety assessment. Current epigenetic studies, including major international sequencing projects, are expected to generate information for establishing the ‘normal’ epigenome of tissues and cell types as well as the physiological variability of the epigenome against which carcinogen exposure can be assessed. Recently, epigenetic events have emerged as key mechanisms in cancer development, and while our search of the Monograph Volume 100 revealed that epigenetics have played a modest role in evaluating human carcinogens by the International Agency for Research on Cancer (IARC) Monographs so far, epigenetic data might play a pivotal role in the future. Here, we review (i) the current status of incorporation of epigenetics in carcinogen evaluation in the IARC Monographs Programme, (ii) potential modes of action for epigenetic carcinogens, (iii) current in vivo and in vitro technologies to detect epigenetic carcinogens, (iv) genomic regions and epigenetic modifications and their biological consequences and (v) critical technological and biological issues in assessment of epigenetic carcinogens. We also discuss the issues related to opportunities and challenges in the application of epigenetic testing in carcinogen identification and evaluation. Although the application of epigenetic assays in carcinogen evaluation is still in its infancy, important data are being generated and valuable scientific resources are being established that should catalyse future applications of epigenetic testing.
Introduction
Epigenetics is a rapidly expanding field of modern biology with a profound impact on our thinking and understanding of biological phenomena. The term ‘epigenetic’ refers to all stable changes in gene expression and chromatin organization that are independent of the DNA sequence itself and that can be mitotically inherited over cell divisions. Epigenetic phenomena, including genomic imprinting, X-chromosome inactivation and global reconfiguration of the DNA methylome, changes in chromatin compaction states and histone modification patterns, occur during development and contribute to the lineage-specific epigenome that is maintained over the lifetime of an organism.
Epigenetic mechanisms are essential for the stable propagation of gene activity states from one generation of cells to the next and thus the epigenome governs the establishment and maintenance of cell identity. DNA methylation, histone modifications and non-coding RNAs are the main epigenetic mechanisms that may act alone or in combination to govern the gene expression programme over the lifetime of an organism.
The remarkable progress in the field of epigenetics has turned academic and medical attention to the potential application of new conceptual advances in cancer research. The advent of novel technologies that allow cost-effective profiling of the epigenome with unprecedented resolution has dramatically accelerated cancer research and opened up new perspectives. Together, these advances have led to a broader appreciation of epigenetics in the a etiology of complex human diseases, including cancer. In addition to their application in mechanistic studies, cancer therapy and biomarker discovery, advances in epigenetics also need to be incorporated into carcinogen identification and safety assessment.
Unlike the genome, which is virtually identical among all cells within an organism, different tissues and cell types harbour a distinct epigenome, which may undergo substantial changes with ageing and in response to environmental factors (1,2). Epigenetic mechanisms can be viewed as an interface between the environment and the genome, the deregulation of which may disrupt key cellular processes, ultimately resulting in oncogenic transformation and tumour development (Figure 1). Exposure to environmental factors may leave a fingerprint on the epigenome that may be exploited in discovering new biomarkers for risk assessment and cancer prevention. Despite an increased interest in epigenetics and a better understanding of epigenetic mechanisms and their deregulation in human malignancies, relatively little is known about the potential application of epigenetics in carcinogen identification and evaluation. Although epigenetic assays have not yet been incorporated into the current carcinogen testing battery, recent evidence on the impact of environmental exposures on the epigenome argues that the time is ripe for established and suspected carcinogens to be specifically evaluated for their potential to deregulate epigenetic mechanisms. The gaps in our understanding of the normal variability of the epigenome in different cell types should be bridged by major international sequencing initiatives enabled by the genome/epigenome technology revolution. Once this is available, it will be possible to identify specific changes in the epigenome associated with exposures to a carcinogen. Several recent reviews on the impact of environmental/lifestyle factors on the epigenome have been published (3–7).
Fig. 1.
Epigenetic mechanisms regulate key cellular processes (such as gene transcription, DNA repair and differentiation) and play critical roles in cellular responses to environmental exposures and endogenous stimuli. Deregulation of epigenetic mechanisms may promote the development of abnormal phenotypes and cancer. There is a crosstalk between epigenetic and genetic changes in the process of cancer development and progression. Given that epigenetic and genetic changes coexist in all cancers, it is often unclear what are the primary and secondary events pertinent to carcinogenesis.
The advances in understanding the normal epigenome should also facilitate developing epigenetic assays in carcinogen testing and using epigenetic data in regulatory processes and policy-making. In this review, we will summarize current status of incorporation of epigenetics in carcinogen evaluation by the International Agency for Research on Cancer (IARC) Monographs Programme. We will also assess selected examples of studies demonstrating the epigenetic effects of specific agents in human carcinogenesis. Finally, we will discuss the issues related to opportunities and challenges in incorporating epigenetics into carcinogen evaluation and future perspectives.
Physiological and pathological changes of the epigenome
An important feature of the epigenome is that it is susceptible to normal variations, that is to say that epigenetic modifications of DNA and histones exhibit variations across different cell types and over time. It is thought that the genome needs to be plastic enough to respond to environmental stressors and endogenous cues (4,8). Such induced changes in the epigenome may be fixed and propagated over cell divisions, resulting in long-term or permanent changes in phenotype. Therefore, in contrast to genotype, modulation of the epigenotype is a physiological and essential process that controls an organism’s response to environmental exposures.
The reconfiguration of the epigenome is particularly evident during embryonic development, cell differentiation and ageing (9–11). A complete erasure and re-establishment of the DNA methylation patterns in early embryonic development is a dynamic, but highly regulated, process that results in a profound reconfiguration of the epigenome (including the DNA methylome but also histone marks) (12) and is believed to be highly susceptible to environmental exposures. The reconfiguration of the methylome during early embryonic development provides a striking example of dynamic changes in the epigenome. In the fertilized egg before the fusion of the pronuclei, DNA methylation levels in the maternal and paternal genomes are heavily methylated and both pronuclei have roughly the same methyl-cytosine content (12,13). Soon after the fusion (within approximately an hour), the paternal genome is efficiently demethylated through an active mechanism (active demethylation), and its demethylation levels remain low during a few mitotic divisions. In contrast, the maternal genome undergoes a passive demethylation during several subsequent mitoses. The genome of the early embryo starts to be remethylated by de novo DNA methylases at implantation, and the establishment of DNA methylation patterns occurs in a tissue-specific manner. It is believed that this process of dramatic demethylation/remethylation represents a window of susceptibility to environmental stressors and that adverse changes in the epigenome during early embryonic development may be at the heart of intrauterine programming of childhood and adult diseases (14). Another stage during embryonic development that may be considered vulnerable is the primordial germ cells (15,16). At this stage, foetal germline is dynamically remodelled in a gender-specific manner and epigenetic modifications (such as DNA methylation marking of imprinted genes) are removed (12,17). However, primordial germ cells can contribute genetic material to the future offspring of the foetus; therefore, epigenetic changes induced by carcinogen exposure during this stage may contribute to transgenerational epigenetic inheritance. It is, therefore, essential to account for these events when evaluating carcinogens.
Despite a better understanding of epigenetic mechanisms and their deregulation in human cancer, much remains unknown about the normal dynamic variations of the epigenome and how to distinguish them from adverse epigenetic changes that pose a health risk. In general, the degree to which agents may promote carcinogenesis through epigenetic mechanisms depends on the amount and duration of exposure. The degree to which an adverse exposure alters the epigenome may also strongly depend on variation in susceptibility to the exposure. Individual susceptibility to a given exposure is likely to depend on the epigenetic make-up that dictates an individual’s response and adaptation mechanisms. Differences in individual susceptibility may be attributed to patterns of DNA methylation, histone modifications and non-coding RNAs, as well as genetic make-up.
Genotoxic, non-genotoxic and epigenetic carcinogens
Epigenetic mechanisms are thought to play important roles in the adaptation and response to environmental exposures, although a clear-cut causal relationship between epigenetic change and specific exposure is often difficult to establish. The reason for this may be 3-fold. First, environmental agents may induce subtle changes and quantitative phenotypic manifestation may be evident only after repetitive or prolonged exposure. Second, environmental exposures are likely to induce global non-specific changes in one layer or multiple layers of the epigenome. Third, there is an important gap in our understanding of the ‘normal’ epigenome in tissues and cell types and the normal variability of the epigenome.
Recent studies have implicated epigenetic mechanisms in carcinogenesis linked to environmental exposures, although there is a paucity of evidence demonstrating molecular mechanisms by which the epigenome is deregulated in response to a specific carcinogen. A wide range of known and suspected carcinogens (including chemical, physical and biological agents) have been associated with changes in the epigenome, and it has been suggested that their mode of action may involve disruption of epigenetic mechanisms (Tables I–IV). The effects of these carcinogens on epigenetic states have been either demonstrated experimentally using different animal and cellular models or inferred from epidemiological studies (4,7). Environmental factors associated with epigenetic deregulation include tobacco smoke, arsenic, cadmium, nickel and ionizing and UV radiation. Different infectious agents such as human hepatitis B virus (HBV), hepatitis C virus, human papillomavirus and the bacterium Helicobacter pylori have been shown to deregulate proliferation, cell division and the gene expression pattern of the host cell via an epigenetic strategy. Among dietary factors, alcohol and fat consumption may act as epigenetic carcinogens.
Table I.
Epigenetic mechanisms and Volume 100A carcinogens
| Human carcinogen | Epigenetic mechanism | Model | References | ||
|---|---|---|---|---|---|
| Human samples | Cell lines | Animal model | |||
| Part A: pharmaceuticals | Tamoxifen | ||||
| DNA methylation | * | (18,19) | |||
| * | (20) | ||||
| * | (21,22) | ||||
| Histone marks | * | (22) | |||
| miRNA | * | (23) | |||
| DES | |||||
| DNA methylation | * | (24) | |||
| * | (24–30) | ||||
| Histone marks | * | (31) | |||
| miRNA | * | (32) | |||
| * | (32) | ||||
| Prostaglandin E2 | |||||
| DNA methylation | * | (33,34) | |||
| * | (33) | ||||
| Histone marks | * | (35) | |||
| miRNA | * | (36,37) | |||
| Hormone therapy | |||||
| DNA methylation | * | (38–40) | |||
| Cyclophosphamide | |||||
| miRNA | * | (41) | |||
| Chromium (VI) compounds | |||||
| Histone marks | * | (42,43) | |||
miRNA, micro-RNA.
Asterisk (*) indicates the model used in a given study.
Table IV.
Epigenetic mechanisms and Volume 100E carcinogens
| Human carcinogen | Epigenetic mechanism | Model | References | ||
|---|---|---|---|---|---|
| Human samples | Cell lines | Animal model | |||
| Lifestyle and diet | |||||
| Tobacco smoking | |||||
| DNA methylation | * | (55,176–185) | |||
| * | (178,179,186–188) | ||||
| Histone marks | * | (189) | |||
| Alcohol consumption | |||||
| DNA methylation | * | (190–193) | |||
| * | (194,195) | ||||
| * | (196–198) | ||||
| Histone marks | * | (194,199) | |||
| * | (141,142,200) | ||||
| miRNA | * | (25,201,202) | |||
Asterisk (*) indicates the model used in a given study.
Table II.
Epigenetic mechanisms and Volume 100B carcinogens
| Human carcinogen | Epigenetic mechanism | Model | References | ||
|---|---|---|---|---|---|
| Human samples | Cell lines | Animal model | |||
| Part B: biological agents | |||||
| HBV | |||||
| DNA methylation | * | (44–55) | |||
| * | (44,46,55–60) | ||||
| Histone marks | * | (61,62) | |||
| * | (61–65) | ||||
| * | (61,62) | ||||
| miRNA | * | (66–71) | |||
| * | (64,66–68,72) | ||||
| * | (72) | ||||
| Hepatitis C virus | |||||
| DNA methylation | * | (50,51,73–75) | |||
| * | (76,77) | ||||
| * | (78) | ||||
| Histone marks | * | (79,80) | |||
| miRNA | * | (71,81–84) | |||
| * | (83,85–89) | ||||
| Human papillomaviruses | |||||
| DNA methylation | * | (90,91) | |||
| * | (90,92) | ||||
| Histone marks | * | (93–98) | |||
| miRNA | * | (99–101) | |||
| Human T-cell lymphotropic virus type 1 | |||||
| DNA methylation | * | (102) | |||
| Histone marks | * | (103–105) | |||
| miRNA | * | (106,107) | |||
| * | (107,108) | ||||
| Epstein–Barr virus | |||||
| DNA methylation | * | (109–118) | |||
| * | (109–112,119–121) | ||||
| Histone marks | * | (119,122–126) | |||
| miRNA | * | (127–129) | |||
| * | (70,127,129–140) | ||||
| * | (129) | ||||
| Helicobacter pylori | |||||
| DNA methylation | * | (61,62,141–153) | |||
| * | (141–143,146,154–156) | ||||
| * | (153,155) | ||||
| Histone marks | * | (154,157–160) | |||
| miRNA | * | (161) | |||
| * | (161) | ||||
| Asterisk (*) indicates the model used in a given study. | |||||
Table III.
Epigenetic mechanisms and Volume 100C–D carcinogens
| Human carcinogen | Epigenetic mechanism | Model | References | ||
|---|---|---|---|---|---|
| Human samples | Cell lines | Animal model | |||
| Part C: arsenic, metals, fibres and dusts and Part D: radiation | |||||
| Arsenic | |||||
| DNA methylation | * | (124–130) | |||
| * | (131–136) | ||||
| * | (131,137,138) | ||||
| Histone marks | * | (139) | |||
| miRNA | * | (140) | |||
| Cadmium | |||||
| DNA methylation | * | (141–144) | |||
| * | (145–148) | ||||
| miRNA | * | (149) | |||
| Nickel | |||||
| DNA methylation | * | (150,151) | |||
| * | (18) | ||||
| Histone marks | * | (152–163) | |||
| * | (163) | ||||
| Beryllium | |||||
| DNA methylation | * | (163) | |||
| Asbestos | |||||
| DNA methylation | * | (164) | |||
| miRNA | * | (165) | |||
| X-radiation | |||||
| DNA methylation | * | (166) | |||
| * | (167–169) | ||||
| miRNA | * | (170) | |||
| * | (171) | ||||
| * | (169) | ||||
| Gamma radiation | |||||
| DNA methylation | * | (172) | |||
| miRNA | * | (170,173,174) | |||
| Smoky coal emissions (for cooking and heating) | |||||
| DNA methylation | * | (175) | |||
| Chromium VI compounds | |||||
| Histone marks | * | (176,177) | |||
| Asterisk (*) indicates the model used in a given study. | |||||
It should be noted that many carcinogens may promote tumour development by inducing both epigenetic changes (aberrant DNA methylation and histone modifications) and genetic alterations (mutations). However, individual genetic polymorphisms and epigenetic make-up (‘epigenetic polymorphisms’) may also play pivotal roles in cellular response to environmental stress and thus may represent a part of an individual’s predisposition to developing cancer. Therefore, individual cancer susceptibility is likely to depend not only on genetic but also on epigenetic make-up. These responses involve the action of diverse cellular machineries such as those involved in DNA repair, carcinogen detoxification, cell cycle control and cell death. The technological advances in epigenomics (high-throughput and genome-wide profiling) will soon allow the identification of entire epigenomes (genome-wide patterns of DNA methylation and histone modifications). This may provide critical information for testing the notion that differences in individual susceptibility may also be attributed to germline epigenetic make-up.
In the literature, the terms ‘non-genotoxic’ and ‘epigenetic’ are sometimes used interchangeably (203). However, not all non-genotoxic carcinogens act by altering epigenetic states (DNA methylation, histone modifications or non-coding RNAs). For example, some non-genotoxic agents, such as dioxin, can act via receptor-mediated pathways (204), which cannot be considered as an epigenetic mechanism. This means that although some epigenetic agents are also genotoxic, not all non-genotoxic agents are epigenetic carcinogens and indeed, as discussed below, agents may act by multiple mechanisms. Therefore, oversimplified classification of carcinogens that does not consider potential contributing effects that are neither genetic nor epigenetic is confusing and should be avoided.
The IARC Monographs Programme
IARC Monographs.
The IARC Monographs Programme identifies environmental and lifestyle factors that are human carcinogens (162). Interdisciplinary Working Groups of expert scientists review published studies and evaluate the strength of the evidence that an agent can increase the risk of human cancer. Since 1971, more than 950 agents have been evaluated, of which more than 400 have been identified as carcinogenic, probably carcinogenic, or possibly carcinogenic to humans. Recently, IARC completed a review of the more than 100 chemicals, occupations, physical agents, biological agents and other agents classified as carcinogenic to humans (205).
Volume 100 of the IARC Monographs reviews all agents classified previously by IARC as carcinogenic to humans (Group 1) and is divided into six parts (A, B, C, D, E and F), each of which describes a distinct class of carcinogens. Our search of the Monograph Volume 100 revealed that only a few chemical agents and nutritional or lifestyle factors evaluated by IARC Working Groups have been considered as epigenetic carcinogens. Diethylstilbestrol (DES), chromium (VI) compounds and ionizing radiation have been classified as carcinogens that may act through epigenomic deregulation. Curiously, for all of these, their impact on the epigenome is considered not as the major mechanism but rather as the secondary mechanism. The lack of focus on epigenetics in relation to the mechanistic data is perhaps surprising considering a wealth of studies demonstrating the impact of many of the Volume 100 carcinogens on epigenetic mechanisms (Tables I–IV). One explanation may lie in the fact that many studies failed to address whether changes in the epigenome were causal or associative to a carcinogenic exposure. In this section, we discuss the specific example of studies demonstrating the epigenetic effects of DES and infectious agents in human carcinogenesis.
DES—an example of an epigenetic carcinogen from the IARC Monographs?
DES is a synthetic oestrogen that was widely used (from the 1940s to the 1970s in the USA) to prevent potential miscarriages (through its stimulation of placental synthesis of oestrogen and progesterone) and for the treatment of symptoms associated with menopause and ovariectomy as well as specific vaginal and vulvar conditions (such as inflammation and dystrophy). DES was also used as a postcoital emergency contraceptive (‘morning-after pill’) and to treat other conditions associated with dysfunctional menstrual cycles and female hypogonadism. It has been estimated that 5–10 million US citizens were treated with DES during pregnancy or were exposed to the drug in utero (206). Although nowadays DES is rarely used (e.g. to treat prostate cancer or specific forms of breast cancer), the consequences of its use are still felt among the treated individuals and their progeny.
DES was evaluated by IARC Working Groups in 1978 and 1987, and was evaluated again for Volume 100 (in 2008); IARC has classified this chemical as a Group 1 carcinogen (205). The IARC Monograph of 1987 states that there is sufficient evidence of a causal association between clear cell adenocarcinoma of the vagina or cervix and prenatal exposure to DES (207). It also states that there is sufficient evidence of a causal relationship between cancer of the breast and the use of DES during pregnancy. The carcinogenicity of DES was established or highly suspected in experimental animals before epidemiological studies confirmed its carcinogenicity in humans. Consistent with its oestrogenic properties, DES was shown to induce various effects on the reproductive system in both mice and humans (206,208,209). In a mouse model, it was shown that prenatal and perinatal exposure to DES produces multiple effects in uterine tissue, including uterine cancer (leiomyomas) (209).
Several studies have investigated the mechanism by which DES exposure promotes carcinogenesis. These include studies of gene expression and DNA methylation states in uterine tissue. Specific changes in gene expression in the uterus of young mice treated neonatally were detected after exposure to DES (208). These alterations were detected in specific genes (fos and lactoferrin) and persisted for weeks, even after treatment cessation. Interestingly, gene expression changes were associated with specific epigenetic changes, namely changes in DNA methylation. For example, the genes that were differentially expressed in DES-treated animals also exhibited abnormal DNA methylation (208,210).
The above results strongly suggest that exposure to DES may have a significant and long-term effect on gene expression through epigenetic mechanisms. However, it should be noted that these studies focused on only a few genes and one epigenetic mechanism (DNA methylation). The impact of DES is unlikely to be limited to a small subset of genes, and epigenome-wide studies have yet to be performed. Indeed, microarray-based transcriptome analysis in both rats and mice has revealed DES-induced changes in expression of a wide range of genes, although whether these changes were accompanied by changes in DNA methylation states or other epigenetic alterations (histone modifications and non-coding RNAs) have been little studied (31,32,211).
Another interesting feature of DES exposure is its impact on cancer incidence in subsequent generations. It has been shown that in addition to an increased cancer susceptibility associated with epigenetic changes in DES-treated parents, an epigenetic mechanism may operate in subsequent generations of mice (the F2 generation) (208). These findings further support the notion that DES-induced carcinogenesis may operate through an epigenetic mechanism, although further studies extending to the F3 generation of exposed animals are needed in order to establish a true transgenerational epigenetic inheritance.
Infectious agents and epigenetic mechanisms of carcinogenesis.
There is growing evidence that different infectious agents may promote carcinogenesis through epigenetic mechanisms. For example, evidence has emerged that infection by HBV, a major risk factor for developing liver cancer, promotes hepatocarcinogenesis by inducing epigenetic changes (212,213). HBV encoded protein X (HBx) acts as an oncogenic transcription factor by affecting the expression of important cellular genes. In HBV infection-associated cancer cells, HBx protein was found to up-regulate expression of the DNA methyltransferase (DNMT) genes and transcriptional silencing of key cellular genes has been attributed to promoter hyper-methylation mediated by DNMTs (214–217). This notion is further supported by the finding that HBx also directly interacts with the de novo methyltransferase DNMT3A, directing their recruitment to specific genes and thus affecting their methylation and silencing (56). Interestingly, HBx was also shown to mediate dissociation of DNMT3A from the promoter of a set of genes, thus resulting in their hypomethylation and transcriptional activation (56). However, chromatin immunoprecipitation experiments failed to find recruitment of HBx on the activated gene promoters suggesting that HBx could facilitate displacement of DNMT through an indirect mechanism.
In addition, a histone deacetylase (HDAC) was found to be a direct interacting partner of HBx protein, revealing a potential alternative epigenetic mechanism for its transcriptional suppressive activities (56). Furthermore, HBx was also shown to directly interact with histone acetyltransferase (HAT) complex CBP/P300 and HBx-mediated recruitment of CBP/P300 complex resulting in hyperacetylation of local chromatin and transactivation of the target cellular genes (44,56,63,218,219). HBx was also found to down-regulate the expression of E-cadherin, the gene frequently found silenced in liver cancer, by the recruitment of the mSin3A/HDAC complex to the E-cadherin gene (64). Therefore, in addition to DNA methylation changes, histone deacetylation of key cellular genes may be an important epigenetic mechanism contributing to HBV-related carcinogenesis. Interestingly, in infected cells, the HBV genome remains in minichromosomes (small chromatin-like structures composed of histones and non-histone proteins and additional genetic material that replicate autonomously) and HBx protein was shown to regulate transcription of both viral genes through epigenetic mechanisms (220–222). However, whether the HBx-mediated engagement of cellular epigenetic machineries in replication and transcription of the viral minichromosomes on critical processes of the host cell remains to be investigated. Although epigenetic mechanisms involved in carcinogenesis associated with viruses other than HBV are less understood, recent studies have suggested that human oncogenic viruses (including Epstein–Barr virus, simian virus 40, Kaposi’s sarcoma-associated herpes virus and hepatitis C virus) may be involved in the deregulation of epigenetic modifiers, ultimately resulting in deregulation of the host genes (6,7,9). Therefore, human oncogenic viruses, in general, may promote carcinogenesis through different epigenetic mechanisms.
Helicobacter pylori is another example of an infectious agent that might promote carcinogenesis through epigenetic mechanisms (223). Helicobacter pylori-infected individuals with chronic gastritis have a significantly higher risk of developing gastric cancer (224) and studies on animal models demonstrated that infiltration of macrophages and expression of inflammation-related genes (proinflammatory cytokines) are associated with DNA methylation changes (225). Further mechanistic studies showed that DNA methylation changes associated with H. pylori-induced chronic inflammation could be suppressed when the infected animals are treated with an anti-inflammatory agent, despite the fact that the presence of H. pylori in gastric mucosae is unaffected (225). These findings argue that the mechanism by which some infectious agents promote carcinogenesis may involve DNA methylation changes induced by inflammation-induced mediators (223,226).
Potential modes of actions for epigenetic carcinogens
Generally, epigenetic carcinogens may promote carcinogenesis (i) through inducing direct changes in the epigenome or (ii) through an indirect deregulation of the epigenetic states. The agents that directly interact with and modulate methyl-cytosine or histone marks may be considered as direct epigenetic carcinogens. Nickel chloride, which was shown to induce changes in histone marks (227), may be considered as a direct epigenetic carcinogen.
The agents in the second group (indirect epigenetic carcinogens) include those that alter either the expression or the activity of enzymes involved in establishing and maintaining epigenetic patterns. The agents that deregulate the activity of de novo DNA methylation (DNMT3A and DNMT3B) or the DNA maintenance methyltransferase (DNMT1) can be considered as indirect epigenetic carcinogens. In addition, this group of agents should include the agents capable of altering the activity of proteins and protein complexes responsible for histone modifications, such as HATs and HDACs. Consistent with the evidence that HATs and histone acetylation are involved in the process of DNA repair (228), the agents that inhibit HAT and HDAC activities may compromise critical cellular processes and consequently compromise genomic stability. Reduced levels of histone acetylation or enhanced histone deacetylation may result in the compaction of chromatin, blocking access of DNA repair factors to DNA lesions. Therefore, epigenetic carcinogens may transiently alter chromatin-modifying/remodelling activities, thus impeding DNA repair and other chromatin-based processes. Consistent with this notion, regional mutation rates in cancer genomes were found to be largely influenced by chromatin organization (229).
Because there is intimate and mutually reinforcing crosstalk between the three epigenetic mechanisms in setting up and maintaining the genome-wide expression programme, epigenetic carcinogens that affect one of the interdependent epigenetic mechanisms are likely to also deregulate the other layers of the epigenome.
Our search of the Monograph Volume 100 revealed that epigenetic deregulation has been considered as a potential mechanism of carcinogenesis for only a handful of carcinogens (Table V). Yet, our comprehensive review of experimental evidence in the literature provided strong arguments for an important role of epigenetic deregulation in the mechanism by which many Group 1 carcinogens may contribute to carcinogenesis (Tables I–IV). This discrepancy may be explained by the fact that most mechanistic data on carcinogen-induced epigenome deregulation have been generated since the Volume 100 evaluation, but also by the need to resolve several issues before epigenetic testing can be fully incorporated into carcinogen evaluation. Less understanding of the relevance of epigenetic alterations and more familiarity with genetic changes as a prevailing mechanism of carcinogenesis may also account for epigenetic changes being overlooked as an important mechanism targeted by human carcinogens. Ongoing and future studies in environmental epigenetics and epigenetic toxicology may prove critical in providing crucial insights into the epigenetic mechanisms by which environmental carcinogens contribute to cancer development and progression.
Table V.
Epigenetic mechanisms and Volume 100 carcinogens
| Agent | Major mechanism | Second mechanism | Human cancer | Comment |
|---|---|---|---|---|
| DES | Oestrogen receptor- dependent pathway | Epigenetic reprogramming | Cervix, vagina, breast, endometrium (limited evidence), testis (limited evidence) | |
| HBV | Integration into host DNA | Hepatocellular carcinoma, cholangiocarcinoma (limited evidence), non- Hodgkin lymphoma (limited evidence) | Epigenetic silencing of tumour suppressor genes; interaction with aflatoxins | |
| Chromium VI compounds | Genotoxicity | Epigenetic effects | Lung, nasal cavity and paranasal sinuses (limited evidence) | |
| Ionizing radiation | Energy transfer in clusters | Epigenetic effects | Many | |
| Coke production | Genotoxicity | Epigenetic effects | Lung |
Current in vivo and in vitro technologies to detect epigenetic carcinogens
Animal models represent an essential tool in carcinogen evaluation, and they are critical for epigenetic testing. However, in addition to the complex nature of the epigenome among species and across different tissues, the lack of appropriate animal models represents the major obstacle in studying the impact of environmental carcinogens on the epigenome. Several animal models have been used to show that exposure to environmental agents has an impact on epigenetic states. These include mouse, rat, rabbit, Drosophila, Caenorhabditis elegans and zebrafish. A few mouse models harbouring locus-specific reporters have been developed and exploited in studying environmentally induced epigenetic changes. For example, the yellow agouti mouse model has been used as a sensitive indicator of locus-specific epigenetic changes, and several studies have demonstrated its utility in studying the role of nutritional modulation on epigenetic states (230–232). The coat colour of the agouti (A vy) mice may be used as a sensitive read-out of locus-specific DNA methylation states (Figure 2). This model proved to be instrumental in investigating the influence of maternal diet during pregnancy on the phenotype of the offspring (231,233). Specifically, it was found that feeding pregnant female mice a diet supplemented with folic acid, vitamin B12 or choline results in noticeable changes in the coat colour of their offspring (231). The loci responsible for the variable phenotype (coat colour) and which are susceptible to modulation by environmental exposures are known as metastable epialleles.
Fig. 2.
Testing the effects of maternal diet on the phenotype of the offspring using the Avy/a mouse model. (A) The effect of dietary supplementation of female mice before or after mating with male Avy/a agouti mice can be tested. (B) The effect of maternal dietary supplementation on the epigenome can be ‘read’ by coat-colour distribution in Avy/a offspring. (C) The effect of DNA methylation at the intracisternal A particle (IAP) on agouti gene expression. Maternal dietary supplementation may shift the coat-colour distribution of the offspring. This shift is a result of change in methylation at IAP element upstream of the agouti gene. Adapted from ref. (234).
In addition to the agouti (A vy) model, another murine metastable epiallele, axin fused [Axin(Fu)] (235), has been shown to exhibit epigenetic plasticity in response to changes in maternal nutrition (236). Furthermore, a model with a new metastable epiallele, CDK5 activator- binding protein intracisternal A particle (Cabp IAP), has been characterized (237). Identification of new metastable epialleles suggests that this mechanism operates at multiple loci across the mouse genome. However, the locus activity and phenotype associated with metastable mouse models are dictated by the DNA methylation pattern at the specific regulatory elements (retrotransposons) of the locus. Therefore, phenotypes associated with modulation of epialleles are locus specific and cannot be easily extrapolated to the rest of the genome.
Despite a wide range of studies that have used these models, their unequivocal utility has been demonstrated only in the context of mother-to-offspring transmission of dietary/environmental cues. The models with metastable epialleles are also likely to contribute to characterizing the mechanism underlying transgenerational epigenetic inheritance. Current and future studies have yet to demonstrate the utility of these models in mechanistic studies aimed at identifying and evaluating carcinogens. More recently, metastable epialleles have been identified in humans (238), and with the completion of major epigenome profiling initiatives, many more metastable epialleles are likely to be identified. However, metastable epialleles are likely to represent a tiny fraction of the epigenome; therefore, should carcinogens modulate these alleles, these changes may not be considered as representative of the entire epigenome.
In addition to the mouse models discussed above, rats can be considered for epigenetic testing of carcinogens. The rat has been extensively characterized for transgenerational inheritance, and there is considerable knowledge of rat embryonic and postnatal development. Therefore, the rat may be a good model for assessing the role of environmentally induced epigenetic deregulation on development and teratogenicity as well as transgenerational epigenetic inheritance (239). However, several drawbacks associated with rat models outweigh these advantages. The genome and epigenome are far better characterized in mice than in rats, and tools are available for analyzing genome-wide changes in epigenetic states in the mouse. Therefore, the mouse represents a highly tractable model for epigenetic carcinogenicity testing. Among other in vivo model systems, C. elegans, Drosophila, zebrafish and honeybees offer the potential to be used in epigenetic evaluation of environmental carcinogens. For example, the fruit fly Drosophila melanogaster, a classic model for genetic research, has recently been suggested as a potential epigenetic model organism (240). In this regard, position effect variegation, the change in phenotype resulting from the change of a gene’s position in the genome, was first discovered through observations of eye colour. Therefore, Drosophila eye colour may serve as an attractive read-out in studying epigenetic deregulation in response to carcinogenic exposure. However, a recent study using genome-scale sequencing at single-base resolution revealed that the genome of Drosophila lacks detectable DNA methylation patterns (241), consistent with the notion that DNA methylation is dispensable for some eukaryotic organisms. Similarly, the nematode worm C. elegans completely lacks genomic DNA methylation (242). Because both C. elegans and Drosophila have been used extensively in developmental biology, these organisms may represent suitable models in high-throughput screening approaches where epigenetic mechanisms (histone modifications and non-coding RNAs) can be studied independent of DNA methylation.
Finally, the use of mammalian cells (human and rodent) grown in culture should also be considered for epigenetic testing. Among these in vitro models, the use of stem cells (embryonic and tissue specific) may prove particularly informative. Cell lines may prove most valuable in untargeted screening and identification of potential epigenetic carcinogens. In vitro models may also be instrumental in a focused dissection of epigenetic events associated with carcinogen exposure, such as molecular pathways analysis and identification of gene targets. Two major shortcomings of cellular models are their susceptibility to epigenetic alterations during in vitro culture and their incompatibility with transgenerational assessment, neither of which are limitations of in vivo models.
Genomic regions and epigenetic modifications and their biological consequences
A distinguishing feature of epigenetic changes that needs to be considered in carcinogen evaluation is their intrinsic reversibility. Therefore, although epigenetic changes are generally stable and are usually transmitted with extreme fidelity over many cell generations, it is possible that adverse changes in DNA methylation, histone modifications and expression of non-coding RNAs associated with carcinogen exposure may be modulated by subsequent exposure to other epigenome-modulating agents.
Although intrinsic to all cells and absolutely essential, the plasticity and reversibility of the epigenome represents an important challenge in the development of methodologies and a battery of assays for epigenetic testing. In principle, epigenetic assays can be developed at two levels: (i) epigenetic patterns or marks and (ii) the expression and/or activity of epigenetic players (enzymes and other molecules involved in setting up and erasing epigenetic marks). Many environmental chemicals are likely to interfere with the activity of epigenetic players, some of which may contribute to carcinogenesis. Although changes in the protein levels and activity of epigenetic machinery players (e.g. DNMT and HDAC enzymes) can be accurately measured, establishing whether the change in the expression or activity represents a lasting adverse effect with phenotypic consequences or merely an adaptive response is more challenging. Similarly, changes in epigenetic patterns (levels and patterns of DNA methylation and histone modification marks) can be detected with increasing ease and accuracy. In both cases, it is critical to establish whether a given change in either of these two epigenetic layers is anchored to a phenotypic outcome. Nevertheless, assays that monitor the epigenetic patterns should be strongly preferred as they are more likely to detect lasting changes in the epigenome.
Another important consideration is that epigenetic changes that are identified in response to a specific carcinogen should be combined with assays that establish a causative or correlative link with adverse phenotype. In this regard, it has been suggested that classifying epigenetic-based phenotypes based on the gene pathway and ontology databases may be a valuable approach (243). For example, the Gene Ontology database that is being developed with the aim of describing genes and pathways in terms of their molecular functions and associated biological processes should be a useful tool in carcinogen evaluation. An important feature of this facility is that it should allow properties to be assigned to genes and pathways in a species-independent manner, although in terms of epigenetic testing an ideal database should be exclusively focused on epigenetic effects and sets of genes and pathways that are known to be influenced by epigenetic mechanisms.
It is well established that epigenetic patterns are species- and tissue specific; however, epigenetic changes are likely to also be exposure specific. Because the epigenome is considered as a cellular mechanism that responds to environmental exposures, many changes in epigenetic states are likely to be a consequence of an adaptive response to adverse exposures. The key question is how to distinguish adaptive from adverse responses in the epigenome. In order to detect adverse epigenetic changes, it is essential to have a comprehensive and detailed database for comparison. With the ongoing international efforts (such as the International Human Epigenome Consortium, http://ihec-epigenomes.net), it is hoped that the epigenomes (DNA methylomes) of a wide range of human and other cell types will be available, as well as the normal variability of the epigenome. The fact that the field of epigenetics is rapidly advancing means that in the near future, we are likely to be in a position to construct a ‘normal’ reference epigenome for comparison and identification of adverse effects of potential carcinogens. Nevertheless, experiments aimed at evaluating the epigenetic effect of a carcinogen should be carefully designed to include the appropriate control group that contains a normal epigenome for comparison.
Critical technological and biological issues in assessment of epigenetic carcinogens
In recent years, we have witnessed an emergence of powerful technologies in epigenetics and epigenomics that allow the sensitive, high-throughput and genome-wide detection of epigenetic changes in normal and cancer cells (244–246). These advances, notably those linked to the development and application of microarrays and massively parallel sequencing technologies, have accelerated epigenomic research and opened up new perspectives. A wide range of methods and approaches exist for the identification, quantification and mapping of changes in the epigenome. Although the earliest approaches were mostly qualitative, typically non-specific and at best useful for quantification of total epigenetic marks in cells, this field has seen considerable progress and development over the past decade.
Methods for DNA methylation analysis differ in their coverage and sensitivity, and the method of choice depends on the intended application and desired level of information. These methods include global methyl-cytosine content, degree of methylation at specific loci or genome-wide methylation maps. With the advent of more advanced and cost-effective technologies, notably DNA microarray platforms and massively parallel sequencing, it is possible to generate comprehensive maps of epigenomes with relative ease. Similarly, a wide range of robust and genome-wide approaches have been developed for analysis of histone modifications and non-coding RNAs. These technological advances will be instrumental in establishing the epigenome in normal and diseased tissues. Considering the intimate crosstalk between the different epigenetic mechanisms, there may be value in designing approaches that aim to interrogate all layers of the epigenome (DNA methylation, histone modifications and non-coding RNA-mediated gene silencing) in response to carcinogen exposure.
Despite the remarkable progress in epigenomics, challenges still remain with regard to the analysis and interpretation of the large data sets generated by the new sequencing platforms. An important challenge will be establishing the ‘normal’ state and the dynamic variation of the epigenome. A comprehensive understanding of the physiological variation of epigenetic states in different cell types will be critical to the capacity to discern between normal and abnormal epigenetic patterns. The development of new bioinformatics tools and epigenetic databases should facilitate these efforts (247).
Despite their genome-wide coverage, high resolution and cost effectiveness, most methods for epigenome analysis are not compatible with the analysis of cell subpopulations. It is particularly problematic when specific epigenetic effects, similar to genetic or other molecular effects, in mixed populations of cells need to be resolved (248). This difficulty is exemplified by the heterogeneous nature of tumour tissues, where the presence of normal (‘contaminating’) cells is common.
In applying epigenetic testing, exposure to carcinogens may induce changes in specific target subpopulations of cells (e.g. stem/progenitor cells) in a tissue. Due to an inability to resolve the epigenome of a single cell and the inevitable averaging of epigenetic data, carcinogen-induced effects may be masked by tissue heterogeneity. Therefore, in addition to the capacity to map the epigenome in great detail, further effort should focus on developing robust methods capable of isolating specific cell fractions and analyzing the epigenome of single cells. Although previously established protocols can be adapted, this area is in need of further development. In addition, significant attention should be paid to selecting appropriate cell populations or subpopulations for epigenetic testing. In this regard, the use of embryonic and tissue-specific stem/progenitor cells should be considered.
Conclusions and perspectives
Unlike the genetic code, which is virtually the same in every single cell of an organism, the epigenetic code shows wide-ranging variability across different cell types and also in the same cells at different developmental stages and under the influence of various environmental stimuli. This plasticity of the epigenetic code poses a significant challenge in epigenetic testing. Current epigenetic studies, including major international sequencing projects, are expected to generate information for establishing the ‘normal’ epigenome of tissues and cell types as well as the physiological variability of the epigenome. This should facilitate studies focusing on individual carcinogens and adverse epigenetic effects associated with carcinogen exposure. It is noteworthy that more broadly than the categories of agents considered to date in the Monographs, other environmental exposures such as obesity, physical inactivity and stress are also likely to act through epigenetic mechanisms.
It is anticipated that chemical compounds may be classified as epigenetic carcinogens based on mechanistic evidence (in a development analogous to that applied to aristolochic acid, which was upgraded to a Group 1 carcinogen based on mechanistic data) (249). Therefore, the mechanistic epigenetic data may be used for non-genotoxic and non-receptor-mediated carcinogens (such as arsenic) or when the carcinogenicity data in humans are inconclusive. Identifying a priority set of potential epigenetic carcinogens [e.g. those classified by IARC as probably carcinogenic or possibly carcinogenic to humans (Groups 2A and 2B) that are known to be non-genotoxic] to be addressed in a systematic way may be a good starting point.
Several issues need to be resolved before epigenetic testing can be fully incorporated into carcinogen identification and evaluation and eventually used in policy decision making. In order to incorporate epigenetic data into carcinogen evaluation, it is necessary to consider which epigenetic marks are evaluated, which assays and model systems are used and how changes in the epigenome are interpreted in terms of their potential to contribute to carcinogenesis. The cellular heterogeneity of tumour and normal tissues represents an important challenge for the interpretation of epigenomic data. Because normal or tumour tissues are rarely composed of an identical cell type or clone, tissue purity needs to be carefully considered for accurately measuring epigenetic changes associated with specific exposures. For example, in analysing and interpreting epigenetic data associated with exposures, one may consider applying a recently developed set of analytical tools (247) for inferring changes in the distribution of different cell subpopulations using DNA methylation signatures, a method that circumvents the need for extensive flow cytometry sorting (250,251).
Next-generation sequencing will allow remarkable accuracy, sensitivity and deep read coverage of epigenetic changes; however, extending this approach to a defined cell subpopulation or to the single-cell level remains a challenge. In addition, almost all epigenetic profiling studies have been focused on identifying epigenetic changes associated with annotated genes. Further efforts aimed at improving our understanding of the functional impact of aberrant epigenetic changes occurring in non-genic regions of the genome will require a comprehensive analytical methodology capable of integrating epigenomic data and transcriptomic data as well as genetic data. The development and exploitation of the Gene Ontology database and the Encyclopedia of DNA Elements (ENCODE; nature.com/encode) should help in this task.
Understanding the epigenetic mechanisms by which epigenetic carcinogens promote cancer development will require bringing together dedicated teams, not only of molecular biologists, toxicologists, pathologists and oncologists but also of bioinformatics and computational experts, in order to establish epigenetic assays and translate epigenomic data into an efficient and systematic evaluation of carcinogens.
Despite these significant challenges, the remarkable advances in epigenetics have provided important insights into the epigenetic mechanisms underlying carcinogenesis. The inclusion of epigenetics in the agenda of the IARC workshops on ‘Tumour Concordance and Mechanisms of Carcinogenesis’, held in April and November 2012, is a testimony to a growing recognition of the importance of epigenetic mechanisms in carcinogenesis. Incorporating epigenetic mechanisms into carcinogen identification and evaluation and risk assessment will be an important legacy of the IARC Monographs Programme.
Funding
PhD fellowship from l’Association pour le Recherche Contre le Cancer (l’ARC to M.-P.L.); European Union (260791, 308610); French National Cancer Institute (INCa).
Acknowledgement
We are grateful to Karen Müller for editing the manuscript.
Glossary
Abbreviations:
- DES
diethylstilbestrol
- DNMT
DNA methyltransferase
- HAT
histone acetyltransferase
- HBV
hepatitis B virus
- HBx
HBV encoded protein X
- HDAC
histone deacetylase
- IARC
International Agency for Research on Cancer.
References
- 1. Bell J.T, et al. (2011) A twin approach to unraveling epigenetics. Trends Genet., 27, 116–125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Christensen B.C, et al. (2009). Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG island context. PLoS Genet., 5, e1000602 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Hou L, et al. (2012) Environmental chemical exposures and human epigenetics. Int. J. Epidemiol., 41, 79–105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Feil R, et al. (2011) Epigenetics and the environment: emerging patterns and implications. Nat. Rev. Genet., 13, 97–109 [DOI] [PubMed] [Google Scholar]
- 5. Baccarelli A, et al. (2009) Epigenetics and environmental chemicals. Curr. Opin. Pediatr., 21, 243–251 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Herceg Z. (2007). Epigenetics and cancer: towards an evaluation of the impact of environmental and dietary factors. Mutagenesis, 22, 91–103 [DOI] [PubMed] [Google Scholar]
- 7. Herceg Z, et al. (2011) Epigenetic mechanisms and cancer: an interface between the environment and the genome. Epigenetics, 6, 804–819 [DOI] [PubMed] [Google Scholar]
- 8. Jaenisch R, et al. (2003). Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat. Genet., 33, 245–254 [DOI] [PubMed] [Google Scholar]
- 9. Milosavljevic A. (2011) Emerging patterns of epigenomic variation. Trends Genet., 27, 242–250 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Biron V.L, et al. (2004) Distinct dynamics and distribution of histone methyl-lysine derivatives in mouse development. Dev. Biol., 276, 337–351 [DOI] [PubMed] [Google Scholar]
- 11. Barouki R, et al. (2012) Developmental origins of non-communicable disease: implications for research and public health. Environ. Health, 11, 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Reik W. (2007). Stability and flexibility of epigenetic gene regulation in mammalian development. Nature, 447, 425–432 [DOI] [PubMed] [Google Scholar]
- 13. Mayer W, et al. (2000). Demethylation of the zygotic paternal genome. Nature, 403, 501–502 [DOI] [PubMed] [Google Scholar]
- 14. Gluckman P.D, et al. (2008). Effect of in utero and early-life conditions on adult health and disease. N. Engl. J. Med., 359, 61–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Skinner M.K. (2011) Environmental epigenetic transgenerational inheritance and somatic epigenetic mitotic stability. Epigenetics, 6, 838–842 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Anway M.D, et al. (2005). Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science, 308, 1466–1469 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Smith Z.D, et al. (2013) DNA methylation: roles in mammalian development. Nat. Rev. Genet., 14, 204–220 [DOI] [PubMed] [Google Scholar]
- 18. Liggett T.E, et al. (2011). Methylation patterns in cell-free plasma DNA reflect removal of the primary tumor and drug treatment of breast cancer patients. Int. J. Cancer, 128, 492–499 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Wu H, et al. (2005). Hypomethylation-linked activation of PAX2 mediates tamoxifen-stimulated endometrial carcinogenesis. Nature, 438, 981–987 [DOI] [PubMed] [Google Scholar]
- 20. Pietkiewicz P.P, et al. (2010). Tamoxifen epigenetically modulates CXCL12 expression in MCF-7 breast cancer cells. Biomed. Pharmacother., 64, 54–57 [DOI] [PubMed] [Google Scholar]
- 21. Pathak S, et al. (2009). Effect of tamoxifen treatment on global and insulin-like growth factor 2-H19 locus-specific DNA methylation in rat spermatozoa and its association with embryo loss. Fertil. Steril., 91, 2253–2263 [DOI] [PubMed] [Google Scholar]
- 22. Tryndyak V.P, et al. (2006). Effect of long-term tamoxifen exposure on genotoxic and epigenetic changes in rat liver: implications for tamoxifen-induced hepatocarcinogenesis. Carcinogenesis, 27, 1713–1720 [DOI] [PubMed] [Google Scholar]
- 23. Pogribny I.P, et al. (2007). Induction of microRNAome deregulation in rat liver by long-term tamoxifen exposure. Mutat. Res., 619, 30–37 [DOI] [PubMed] [Google Scholar]
- 24. Bromer J.G, et al. (2009). Hypermethylation of homeobox A10 by in utero diethylstilbestrol exposure: an epigenetic mechanism for altered developmental programming. Endocrinology, 150, 3376–3382 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Tang W.Y, et al. (2008). Persistent hypomethylation in the promoter of nucleosomal binding protein 1 (Nsbp1) correlates with overexpression of Nsbp1 in mouse uteri neonatally exposed to diethylstilbestrol or genistein. Endocrinology, 149, 5922–5931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Li S, et al. (2003). Neonatal diethylstilbestrol exposure induces persistent elevation of c-fos expression and hypomethylation in its exon-4 in mouse uterus. Mol. Carcinog., 38, 78–84 [DOI] [PubMed] [Google Scholar]
- 27. Sato K, et al. (2006). Neonatal exposure to diethylstilbestrol alters the expression of DNA methyltransferases and methylation of genomic DNA in the epididymis of mice. Endocr. J., 53, 331–337 [DOI] [PubMed] [Google Scholar]
- 28. Sato K, et al. (2009). Neonatal exposure to diethylstilbestrol alters expression of DNA methyltransferases and methylation of genomic DNA in the mouse uterus. Endocr. J., 56, 131–139 [DOI] [PubMed] [Google Scholar]
- 29. Li S, et al. (2001). Promoter CpG methylation of Hox-a10 and Hox-a11 in mouse uterus not altered upon neonatal diethylstilbestrol exposure. Mol. Carcinog., 32, 213–219 [DOI] [PubMed] [Google Scholar]
- 30. Alworth L.C, et al. (2002). Uterine responsiveness to estradiol and DNA methylation are altered by fetal exposure to diethylstilbestrol and methoxychlor in CD-1 mice: effects of low versus high doses. Toxicol. Appl. Pharmacol., 183, 10–22 [DOI] [PubMed] [Google Scholar]
- 31. Warita K, et al. (2010). Direct effects of diethylstilbestrol on the gene expression of the cholesterol side-chain cleavage enzyme (P450scc) in testicular Leydig cells. Life Sci., 87, 281–285 [DOI] [PubMed] [Google Scholar]
- 32. Hsu P.Y, et al. (2009). Xenoestrogen-induced epigenetic repression of microRNA-9-3 in breast epithelial cells. Cancer Res., 69, 5936–5945 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Xia D, et al. (2012). Prostaglandin E2 promotes intestinal tumor growth via DNA methylation. Nat. Med., 18, 224–226 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Huang S.K, et al. (2012). Prostaglandin E(2) increases fibroblast gene-specific and global DNA methylation via increased DNA methyltransferase expression. FASEB J., 26, 3703–3714 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Kawagoe J, et al. (2007). Mechanism of the divergent effects of estrogen on the cell proliferation of human umbilical endothelial versus aortic smooth muscle cells. Endocrinology, 148, 6092–6099 [DOI] [PubMed] [Google Scholar]
- 36. Wickramasinghe N.S, et al. (2009). Estradiol downregulates miR-21 expression and increases miR-21 target gene expression in MCF-7 breast cancer cells. Nucleic Acids Res., 37, 2584–2595 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Maillot G, et al. (2009). Widespread estrogen-dependent repression of micrornas involved in breast tumor cell growth. Cancer Res., 69, 8332–8340 [DOI] [PubMed] [Google Scholar]
- 38. Wu A.H, et al. (2010). Hormone therapy, DNA methylation and colon cancer. Carcinogenesis, 31, 1060–1067 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Friso S, et al. (2007). Oestrogen replacement therapy reduces total plasma homocysteine and enhances genomic DNA methylation in postmenopausal women. Br. J. Nutr., 97, 617–621 [DOI] [PubMed] [Google Scholar]
- 40. Cappelletti V, et al. (2008). Patterns and changes in gene expression following neo-adjuvant anti-estrogen treatment in estrogen receptor-positive breast cancer. Endocr. Relat. Cancer, 15, 439–449 [DOI] [PubMed] [Google Scholar]
- 41. Gueta K, et al. (2010). Teratogen-induced alterations in microRNA-34, microRNA-125b and microRNA-155 expression: correlation with embryonic p53 genotype and limb phenotype. BMC Dev. Biol., 10, 20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Xia B, et al. (2011). Chromium(VI) causes down regulation of biotinidase in human bronchial epithelial cells by modifications of histone acetylation. Toxicol. Lett., 205, 140–145 [DOI] [PubMed] [Google Scholar]
- 43. Sun H, et al. (2009). Modulation of histone methylation and MLH1 gene silencing by hexavalent chromium. Toxicol. Appl. Pharmacol., 237, 258–266 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Park I.Y, et al. (2007). Aberrant epigenetic modifications in hepatocarcinogenesis induced by hepatitis B virus X protein. Gastroenterology, 132, 1476–1494 [DOI] [PubMed] [Google Scholar]
- 45. Lambert M.P, et al. (2011). Aberrant DNA methylation distinguishes hepatocellular carcinoma associated with HBV and HCV infection and alcohol intake. J. Hepatol., 54, 705–715 [DOI] [PubMed] [Google Scholar]
- 46. Vivekanandan P, et al. (2010). Hepatitis B virus replication induces methylation of both host and viral DNA. J. Virol., 84, 4321–4329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Oh B.K, et al. (2007). DNA methyltransferase expression and DNA methylation in human hepatocellular carcinoma and their clinicopathological correlation. Int. J. Mol. Med., 20, 65–73 [PubMed] [Google Scholar]
- 48. Zhu Y.Z, et al. (2010). Hepatitis B virus X protein induces hypermethylation of p16(INK4A) promoter via DNA methyltransferases in the early stage of HBV-associated hepatocarcinogenesis. J. Viral Hepat., 17, 98–107 [DOI] [PubMed] [Google Scholar]
- 49. Zhu Y.Z, et al. (2010). Hepatitis B virus X protein promotes hypermethylation of p16(INK4A) promoter through upregulation of DNA methyltransferases in hepatocarcinogenesis. Exp. Mol. Pathol., 89, 268–275 [DOI] [PubMed] [Google Scholar]
- 50. Hernandez-Vargas H, et al. (2010). Hepatocellular carcinoma displays distinct DNA methylation signatures with potential as clinical predictors. PLoS One, 5, e9749 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Feng Q, et al. (2010). DNA methylation changes in normal liver tissues and hepatocellular carcinoma with different viral infection. Exp. Mol. Pathol., 88, 287–292 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Su P.F, et al. (2007). Differential DNA methylation associated with hepatitis B virus infection in hepatocellular carcinoma. Int. J. Cancer, 121, 1257–1264 [DOI] [PubMed] [Google Scholar]
- 53. He Y, et al. (2006). Not polymorphism but methylation of class II transactivator gene promoter IV associated with persistent HBV infection. J. Clin. Virol., 37, 282–286 [DOI] [PubMed] [Google Scholar]
- 54. Jicai Z, et al. (2006). Persistent infection of hepatitis B virus is involved in high rate of p16 methylation in hepatocellular carcinoma. Mol. Carcinog., 45, 530–536 [DOI] [PubMed] [Google Scholar]
- 55. Liu J, et al. (2006). Downregulation of E-cadherin by hepatitis B virus X antigen in hepatocellul ar carcinoma. Oncogene, 25, 1008–1017 [DOI] [PubMed] [Google Scholar]
- 56. Zheng D.L, et al. (2009). Epigenetic modification induced by hepatitis B virus X protein via interaction with de novo DNA methyltransferase DNMT3A. J. Hepatol., 50, 377–387 [DOI] [PubMed] [Google Scholar]
- 57. Niu D, et al. (2009). HBx genotype D represses GSTP1 expression and increases the oxidative level and apoptosis in HepG2 cells. Mol. Oncol., 3, 67–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Jung J.K, et al. (2007). Expression of DNA methyltransferase 1 is activated by hepatitis B virus X protein via a regulatory circuit involving the p16INK4a-cyclin D1-CDK 4/6-pRb-E2F1 pathway. Cancer Res., 67, 5771–5778 [DOI] [PubMed] [Google Scholar]
- 59. Jung J.K, et al. (2010). Hepatitis B virus X protein overcomes the growth-inhibitory potential of retinoic acid by downregulating retinoic acid receptor-beta2 expression via DNA methylation. J. Gen. Virol., 91, 493–500 [DOI] [PubMed] [Google Scholar]
- 60. Lee J.O, et al. (2005). Hepatitis B virus X protein represses E-cadherin expression via activation of DNA methyltransferase 1. Oncogene, 24, 6617–6625 [DOI] [PubMed] [Google Scholar]
- 61. Yoo E.J, et al. (2008). Helicobacter pylori-infection-associated CpG island hypermethylation in the stomach and its possible association with polycomb repressive marks. Virchows Arch., 452, 515–524 [DOI] [PubMed] [Google Scholar]
- 62. Yoo Y.G, et al. (2008). Hepatitis B virus X protein induces the expression of MTA1 and HDAC1, which enhances hypoxia signaling in hepatocellular carcinoma cells. Oncogene, 27, 3405–3413 [DOI] [PubMed] [Google Scholar]
- 63. Shon J.K, et al. (2009). Hepatitis B virus-X protein recruits histone deacetylase 1 to repress insulin-like growth factor binding protein 3 transcription. Virus Res., 139, 14–21 [DOI] [PubMed] [Google Scholar]
- 64. Arzumanyan A, et al. (2012). Epigenetic repression of E-cadherin expression by hepatitis B virus x antigen in liver cancer. Oncogene, 31, 563–572 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Yang L, et al. (2009). Hepatitis B virus X protein upregulates expression of SMYD3 and C-MYC in HepG2 cells. Med. Oncol., 26, 445–451 [DOI] [PubMed] [Google Scholar]
- 66. Zhang J, et al. (2011). Methylation of RAR-β2, RASSF1A, and CDKN2A genes induced by nickel subsulfide and nickel-carcinogenesis in rats. Biomed. Environ. Sci., 24, 163–171 [DOI] [PubMed] [Google Scholar]
- 67. Zhang X, et al. (2011). Modulation of hepatitis B virus replication and hepatocyte differentiation by microRNA-1. Hepatology, 53, 1476–1485 [DOI] [PubMed] [Google Scholar]
- 68. Zhang Z.Z, et al. (2011). Hepatitis B virus and hepatocellular carcinoma at the miRNA level. World J. Gastroenterol., 17, 3353–3358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Li G, et al. (2010). MicroRNA-10b induced by Epstein-Barr virus-encoded latent membrane protein-1 promotes the metastasis of human nasopharyngeal carcinoma cells. Cancer Lett., 299, 29–36 [DOI] [PubMed] [Google Scholar]
- 70. Li L.M, et al. (2010). Serum microRNA profiles serve as novel biomarkers for HBV infection and diagnosis of HBV-positive hepatocarcinoma. Cancer Res., 70, 9798–9807 [DOI] [PubMed] [Google Scholar]
- 71. Ura S, et al. (2009). Differential microRNA expression between hepatitis B and hepatitis C leading disease progression to hepatocellular carcinoma. Hepatology, 49, 1098–1112 [DOI] [PubMed] [Google Scholar]
- 72. Kong G, et al. (2011). Upregulated microRNA-29a by hepatitis B virus X protein enhances hepatoma cell migration by targeting PTEN in cell culture model. PLoS One, 6, e19518 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Deng Y.B, et al. (2010). Identification of genes preferentially methylated in hepatitis C virus-related hepatocellular carcinoma. Cancer Sci., 101, 1501–1510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Lu C.Y, et al. (2009). Aberrant DNA methylation profile and frequent methylation of KLK10 and OXGR1 genes in hepatocellular carcinoma. Genes Chromosomes Cancer, 48, 1057–1068 [DOI] [PubMed] [Google Scholar]
- 75. Nishida N, et al. (2008). Aberrant methylation of multiple tumor suppressor genes in aging liver, chronic hepatitis, and hepatocellular carcinoma. Hepatology, 47, 908–918 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Lim J.S, et al. (2012). Hepatitis C virus core protein overcomes stress-induced premature senescence by down-regulating p16 expression via DNA methylation. Cancer Lett., 321, 154–161 [DOI] [PubMed] [Google Scholar]
- 77. Ripoli M, et al. (2011). Hypermethylated levels of E-cadherin promoter in Huh-7 cells expressing the HCV core protein. Virus Res., 160, 74–81 [DOI] [PubMed] [Google Scholar]
- 78. Higgs M.R, et al. (2010). Downregulation of Gadd45beta expression by hepatitis C virus leads to defective cell cycle arrest. Cancer Res., 70, 4901–4911 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Miura K, et al. (2008). Hepatitis C virus-induced oxidative stress suppresses hepcidin expression through increased histone deacetylase activity. Hepatology, 48, 1420–1429 [DOI] [PubMed] [Google Scholar]
- 80. Borowski P, et al. (1999). Identification and characterization of a histone binding site of the non-structural protein 3 of hepatitis C virus. J. Clin. Virol., 13, 61–69 [DOI] [PubMed] [Google Scholar]
- 81. Peveling-Oberhag J, et al. (2012). Dysregulation of global microRNA expression in splenic marginal zone lymphoma and influence of chronic hepatitis C virus infection. Leukemia, 26, 1654–1662 [DOI] [PubMed] [Google Scholar]
- 82. Abdalla M.A, et al. (2012). Promising candidate urinary microRNA biomarkers for the early detection of hepatocellular carcinoma among high-risk hepatitis C virus Egyptian patients. J. Cancer, 3, 19–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Marquez R.T, et al. (2010). Correlation between microRNA expression levels and clinical parameters associated with chronic hepatitis C viral infection in humans. Lab. Invest., 90, 1727–1736 [DOI] [PubMed] [Google Scholar]
- 84. Peng X, et al. (2009). Computational identification of hepatitis C virus associated microRNA-mRNA regulatory modules in human livers. BMC Genomics, 10, 373 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Cermelli S, et al. (2011). Circulating microRNAs in patients with chronic hepatitis C and non-alcoholic fatty liver disease. PLoS One, 6, e23937 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Ishida H, et al. (2011). Alterations in microRNA expression profile in HCV-infected hepatoma cells: involvement of miR-491 in regulation of HCV replication via the PI3 kinase/Akt pathway. Biochem. Biophys. Res. Commun., 412, 92–97 [DOI] [PubMed] [Google Scholar]
- 87. Steuerwald N.M, et al. (2010). Parallel microRNA and mRNA expression profiling of (genotype 1b) human hepatoma cells expressing hepatitis C virus. Liver Int., 30, 1490–1504 [DOI] [PubMed] [Google Scholar]
- 88. Braconi C, et al. (2010). Hepatitis C virus proteins modulate microRNA expression and chemosensitivity in malignant hepatocytes. Clin. Cancer Res., 16, 957–966 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Liu X, et al. (2010). Systematic identification of microRNA and messenger RNA profiles in hepatitis C virus-infected human hepatoma cells. Virology, 398, 57–67 [DOI] [PubMed] [Google Scholar]
- 90. Sartor M.A, et al. (2011). Genome-wide methylation and expression differences in HPV(+) and HPV(− ) squamous cell carcinoma cell lines are consistent with divergent mechanisms of carcinogenesis. Epigenetics, 6, 777–787 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Weiss D, et al. (2011). Promoter methylation of cyclin A1 is associated with human papillomavirus 16 induced head and neck squamous cell carcinoma independently of p53 mutation. Mol. Carcinog., 50, 680–688 [DOI] [PubMed] [Google Scholar]
- 92. Jiang J, et al. (2012). Hypomethylated CpG around the transcription start site enables TERT expression and HPV16 E6 regulates TERT methylation in cervical cancer cells. Gynecol. Oncol., 124, 534–541 [DOI] [PubMed] [Google Scholar]
- 93. Hyland P.L, et al. (2011). Evidence for alteration of EZH2, BMI1, and KDM6A and epigenetic reprogramming in human papillomavirus type 16 E6/E7-expressing keratinocytes. J. Virol., 85, 10999–11006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. McLaughlin-Drubin M.E, et al. (2011). Human papillomavirus E7 oncoprotein induces KDM6A and KDM6B histone demethylase expression and causes epigenetic reprogramming. Proc. Natl Acad. Sci. U S A, 108, 2130–2135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Bodily J.M, et al. (2011). Human papillomavirus E7 enhances hypoxia-inducible factor 1-mediated transcription by inhibiting binding of histone deacetylases. Cancer Res., 71, 1187–1195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Longworth M.S, et al. (2005). HPV31 E7 facilitates replication by activating E2F2 transcription through its interaction with HDACs. EMBO J., 24, 1821–1830 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Avvakumov N, et al. (2003). Interaction of the HPV E7 proteins with the pCAF acetyltransferase. Oncogene, 22, 3833–3841 [DOI] [PubMed] [Google Scholar]
- 98. Hsu C.H, et al. (2012). The HPV E6 oncoprotein targets histone methyltransferases for modulating specific gene transcription. Oncogene, 31, 2335–2349 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Dreher A, et al. (2011). Differential expression of cellular microRNAs in HPV 11, -16, and -45 transfected cells. Biochem. Biophys. Res. Commun., 412, 20–25 [DOI] [PubMed] [Google Scholar]
- 100. Greco D, et al. (2011). Human papillomavirus 16 E5 modulates the expression of host microRNAs. PLoS One, 6, e21646 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Martinez I, et al. (2008). Human papillomavirus type 16 reduces the expression of microRNA-218 in cervical carcinoma cells. Oncogene, 27, 2575–2582 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Nakase K, et al. (2009). Mechanisms of SHP-1 P2 promoter regulation in hematopoietic cells and its silencing in HTLV-1-transformed T cells. J. Leukoc. Biol., 85, 165–174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Bogenberger J.M, et al. (2008). Human T lymphotropic virus type 1 protein Tax reduces histone levels. Retrovirology, 5, 9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Cheng J, et al. (2007). Negative regulation of the SH2-homology containing protein-tyrosine phosphatase-1 (SHP-1) P2 promoter by the HTLV-1 Tax oncoprotein. Blood, 110, 2110–2120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Lu H, et al. (2004). Tax relieves transcriptional repression by promoting histone deacetylase 1 release from the human T-cell leukemia virus type 1 long terminal repeat. J. Virol., 78, 6735–6743 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Bellon M, et al. (2009). Deregulation of microRNA involved in hematopoiesis and the immune response in HTLV-I adult T-cell leukemia. Blood, 113, 4914–4917 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Yeung M.L, et al. (2008). Roles for microRNAs, miR-93 and miR-130b, and tumor protein 53-induced nuclear protein 1 tumor suppressor in cell growth dysregulation by human T-cell lymphotrophic virus 1. Cancer Res., 68, 8976–8985 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Pichler K, et al. (2008). MicroRNA miR-146a and further oncogenesis-related cellular microRNAs are dysregulated in HTLV-1-transformed T lymphocytes. Retrovirology, 5, 100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Matsusaka K, et al. (2011). Classification of Epstein-Barr virus-positive gastric cancers by definition of DNA methylation epigenotypes. Cancer Res., 71, 7187–7197 [DOI] [PubMed] [Google Scholar]
- 110. Paschos K, et al. (2009). Epstein-Barr virus latency in B cells leads to epigenetic repression and CpG methylation of the tumour suppressor gene Bim. PLoS Pathog., 5, e1000492 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Hino R, et al. (2009). Activation of DNA methyltransferase 1 by EBV latent membrane protein 2A leads to promoter hypermethylation of PTEN gene in gastric carcinoma. Cancer Res., 69, 2766–2774 [DOI] [PubMed] [Google Scholar]
- 112. Tsai C.L, et al. (2006). Activation of DNA methyltransferase 1 by EBV LMP1 involves c-Jun NH(2)-terminal kinase signaling. Cancer Res., 66, 11668–11676 [DOI] [PubMed] [Google Scholar]
- 113. Ushiku T, et al. (2007). p73 gene promoter methylation in Epstein-Barr virus-associated gastric carcinoma. Int. J. Cancer, 120, 60–66 [DOI] [PubMed] [Google Scholar]
- 114. Chang M.S, et al. (2006). CpG island methylation status in gastric carcinoma with and without infection of Epstein-Barr virus. Clin. Cancer Res., 12, 2995–3002 [DOI] [PubMed] [Google Scholar]
- 115. Kim J, et al. (2005). Silencing and CpG island methylation of GSTP1 is rare in ordinary gastric carcinomas but common in Epstein-Barr virus-associated gastric carcinomas. Anticancer Res., 25, 4013–4019 [PubMed] [Google Scholar]
- 116. Sakuma K, et al. (2004). High-density methylation of p14ARF and p16INK4A in Epstein-Barr virus-associated gastric carcinoma. Int. J. Cancer, 112, 273–278 [DOI] [PubMed] [Google Scholar]
- 117. Sudo M, et al. (2004). Promoter hypermethylation of E-cadherin and its abnormal expression in Epstein-Barr virus-associated gastric carcinoma. Int. J. Cancer, 109, 194–199 [DOI] [PubMed] [Google Scholar]
- 118. Kang G.H, et al. (2002). Epstein-Barr virus-positive gastric carcinoma demonstrates frequent aberrant methylation of multiple genes and constitutes CpG island methylator phenotype-positive gastric carcinoma. Am. J. Pathol., 160, 787–794 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Skalska L, et al. (2010). Epigenetic repression of p16(INK4A) by latent Epstein-Barr virus requires the interaction of EBNA3A and EBNA3C with CtBP. PLoS Pathog., 6, e1000951 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Grafodatskaya D, et al. (2010). EBV transformation and cell culturing destabilizes DNA methylation in human lymphoblastoid cell lines. Genomics, 95, 73–83 [DOI] [PubMed] [Google Scholar]
- 121. Seo S.Y, et al. (2008). Epstein-Barr virus latent membrane protein 1 suppresses the growth-inhibitory effect of retinoic acid by inhibiting retinoic acid receptor-beta2 expression via DNA methylation. Cancer Lett., 270, 66–76 [DOI] [PubMed] [Google Scholar]
- 122. Maruo S, et al. (2011). Epstein-Barr virus nuclear antigens 3C and 3A maintain lymphoblastoid cell growth by repressing p16INK4A and p14ARF expression. Proc. Natl Acad. Sci. U S A, 108, 1919–1924 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Anderton J.A, et al. (2011). The H3K27me3 demethylase, KDM6B, is induced by Epstein-Barr virus and over-expressed in Hodgkin’s lymphoma. Oncogene, 30, 2037–2043 [DOI] [PubMed] [Google Scholar]
- 124. Knight J.S, et al. (2003). Epstein-Barr virus nuclear antigen 3C recruits histone deacetylase activity and associates with the corepressors mSin3A and NCoR in human B-cell lines. J. Virol., 77, 4261–4272 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Cotter M.A., 2nd, et al. (2000). Modulation of histone acetyltransferase activity through interaction of Epstein-Barr nuclear antigen 3C with prothymosin alpha. Mol. Cell. Biol., 20, 5722–5735 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Radkov S.A, et al. (1999). Epstein-Barr virus nuclear antigen 3C interacts with histone deacetylase to repress transcription. J. Virol., 73, 5688–5697 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Shinozaki A, et al. (2010). Downregulation of microRNA-200 in EBV-associated gastric carcinoma. Cancer Res., 70, 4719–4727 [DOI] [PubMed] [Google Scholar]
- 128. Cosmopoulos K, et al. (2009). Comprehensive profiling of Epstein-Barr virus microRNAs in nasopharyngeal carcinoma. J. Virol., 83, 2357–2367 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Kim D.N, et al. (2007). Expression of viral microRNAs in Epstein-Barr virus-associated gastric carcinoma. J. Virol., 81, 1033–1036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Yu H, et al. (2012). Epstein-Barr virus downregulates microRNA 203 through the oncoprotein latent membrane protein 1: a contribution to increased tumor incidence in epithelial cells. J. Virol., 86, 3088–3099 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Feederle R, et al. (2011). A viral microRNA cluster strongly potentiates the transforming properties of a human herpesvirus. PLoS Pathog., 7, e1001294 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Du Z.M, et al. (2011). Upregulation of MiR-155 in nasopharyngeal carcinoma is partly driven by LMP1 and LMP2A and downregulates a negative prognostic marker JMJD1A. PLoS One, 6, e19137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Imig J, et al. (2011). MicroRNA profiling in Epstein-Barr virus-associated B-cell lymphoma. Nucleic Acids Res., 39, 1880–1893 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Anastasiadou E, et al. (2010). Epstein-Barr virus encoded LMP1 downregulates TCL1 oncogene through miR-29b. Oncogene, 29, 1316–1328 [DOI] [PubMed] [Google Scholar]
- 135. Godshalk S.E, et al. (2008). Epstein-Barr virus-mediated dysregulation of human microRNA expression. Cell Cycle, 7, 3595–3600 [DOI] [PubMed] [Google Scholar]
- 136. Cameron J.E, et al. (2008). Epstein-Barr virus growth/latency III program alters cellular microRNA expression. Virology, 382, 257–266 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Gatto G, et al. (2008). Epstein-Barr virus latent membrane protein 1 trans-activates miR-155 transcription through the NF-kappaB pathway. Nucleic Acids Res., 36, 6608–6619 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Rahadiani N, et al. (2008). Latent membrane protein-1 of Epstein-Barr virus induces the expression of B-cell integration cluster, a precursor form of microRNA-155, in B lymphoma cell lines. Biochem. Biophys. Res. Commun., 377, 579–583 [DOI] [PubMed] [Google Scholar]
- 139. Motsch N, et al. (2007). Epstein-Barr virus-encoded latent membrane protein 1 (LMP1) induces the expression of the cellular microRNA miR-146a. RNA Biol., 4, 131–137 [DOI] [PubMed] [Google Scholar]
- 140. Lu F, et al. (2008). Epstein-Barr virus-induced miR-155 attenuates NF-kappaB signaling and stabilizes latent virus persistence. J. Virol., 82, 10436–10443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Guo W, et al. (2011). Alcohol exposure decreases CREB binding protein expression and histone acetylation in the developing cerebellum. PLoS One, 6, e19351 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Guo X.B, et al. (2011). Helicobacter pylori induces promoter hypermethylation and downregulates gene expression of IRX1 transcription factor on human gastric mucosa. J. Gastroenterol. Hepatol., 26, 1685–1690 [DOI] [PubMed] [Google Scholar]
- 143. Yan J, et al. (2011). Helicobacter pylori infection promotes methylation of WWOX gene in human gastric cancer. Biochem. Biophys. Res. Commun., 408, 99–102 [DOI] [PubMed] [Google Scholar]
- 144. Yoshida T, et al. (2011) Alu and Satα hypomethylation in Helicobacter pylori-infected gastric mucosae. Int. J. Cancer, 128, 33–39 [DOI] [PubMed] [Google Scholar]
- 145. Shin C.M, et al. (2010). Role of Helicobacter pylori infection in aberrant DNA methylation along multistep gastric carcinogenesis. Cancer Sci., 101, 1337–1346 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Niwa T, et al. (2010). Inflammatory processes triggered by Helicobacter pylori infection cause aberrant DNA methylation in gastric epithelial cells. Cancer Res., 70, 1430–1440 [DOI] [PubMed] [Google Scholar]
- 147. Sepulveda A.R, et al. (2010). CpG methylation and reduced expression of O6-methylguanine DNA methyltransferase is associated with Helicobacter pylori infection. Gastroenterology, 138, 1836–1844 [DOI] [PubMed] [Google Scholar]
- 148. Kondo T, et al. (2009). Accumulation of aberrant CpG hypermethylation by Helicobacter pylori infection promotes development and progression of gastric MALT lymphoma. Int. J. Oncol., 35, 547–557 [DOI] [PubMed] [Google Scholar]
- 149. Nakajima T, et al. (2009). The presence of a methylation fingerprint of Helicobacter pylori infection in human gastric mucosae. Int. J. Cancer, 124, 905–910 [DOI] [PubMed] [Google Scholar]
- 150. Dong C.X, et al. (2009). Promoter methylation of p16 associated with Helicobacter pylori infection in precancerous gastric lesions: a population-based study. Int. J. Cancer, 124, 434–439 [DOI] [PubMed] [Google Scholar]
- 151. Leung W.K, et al. (2006). Effects of Helicobacter pylori eradication on methylation status of E-cadherin gene in noncancerous stomach. Clin. Cancer Res., 12, 3216–3221 [DOI] [PubMed] [Google Scholar]
- 152. Chan A.O, et al. (2003). Promoter methylation of E-cadherin gene in gastric mucosa associated with Helicobacter pylori infection and in gastric cancer. Gut., 52, 502–506 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Peterson A.J, et al. (2010). Helicobacter pylori infection promotes methylation and silencing of trefoil factor 2, leading to gastric tumor development in mice and humans. Gastroenterology, 139, 2005–2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Pero R, et al. (2011). Chromatin and DNA methylation dynamics of Helicobacter pylori-induced COX-2 activation. Int. J. Med. Microbiol., 301, 140–149 [DOI] [PubMed] [Google Scholar]
- 155. Bussière F.I, et al. (2010). H. pylori-induced promoter hypermethylation downregulates USF1 and USF2 transcription factor gene expression. Cell. Microbiol., 12, 1124–1133 [DOI] [PubMed] [Google Scholar]
- 156. Katayama Y, et al. (2009). Helicobacter pylori causes runx3 gene methylation and its loss of expression in gastric epithelial cells, which is mediated by nitric oxide produced by macrophages. Biochem. Biophys. Res. Commun., 388, 496–500 [DOI] [PubMed] [Google Scholar]
- 157. Ding S.Z, et al. (2010). Helicobacter pylori-induced histone modification, associated gene expression in gastric epithelial cells, and its implication in pathogenesis. PLoS One, 5, e9875 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Fehri L.F, et al. (2009). Helicobacter pylori-induced modification of the histone H3 phosphorylation status in gastric epithelial cells reflects its impact on cell cycle regulation. Epigenetics., 4, 577–586 [DOI] [PubMed] [Google Scholar]
- 159. Xia G, et al. (2008). Helicobacter pylori regulates p21(WAF1) by histone H4 acetylation. Biochem. Biophys. Res. Commun., 369, 526–531 [DOI] [PubMed] [Google Scholar]
- 160. Angrisano T, et al. (2012). Helicobacter pylori regulates iNOS promoter by histone modifications in human gastric epithelial cells. Med. Microbiol. Immunol., 201, 249–257 [DOI] [PubMed] [Google Scholar]
- 161. Matsushima K, et al. (2011). MicroRNA signatures in Helicobacter pylori-infected gastric mucosa. Int. J. Cancer, 128, 361–370 [DOI] [PubMed] [Google Scholar]
- 162. Cogliano V.J, et al. (2011) Preventable exposures associated with human cancers. J. Natl Cancer Inst., 103, 1827–1839 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Belinsky S.A, et al. (2002). Aberrant CpG island methylation of the p16(INK4a) and estrogen receptor genes in rat lung tumors induced by particulate carcinogens. Carcinogenesis, 23, 335–339 [DOI] [PubMed] [Google Scholar]
- 164. Christensen B.C, et al. (2008). Asbestos exposure predicts cell cycle control gene promoter methylation in pleural mesothelioma. Carcinogenesis, 29, 1555–1559 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Nymark P, et al. (2011). Integrative analysis of microRNA, mRNA and aCGH data reveals asbestos- and histology-related changes in lung cancer. Genes Chromosomes Cancer, 50, 585–597 [DOI] [PubMed] [Google Scholar]
- 166. Krakowczyk L, et al. (2010). Effects of X-ray irradiation on methylation levels of p16, MGMT and APC genes in sporadic colorectal carcinoma and corresponding normal colonic mucosa. Med. Sci. Monit., 16, CR469–CR474 [PubMed] [Google Scholar]
- 167. Giotopoulos G, et al. (2006). DNA methylation during mouse hemopoietic differentiation and radiation-induced leukemia. Exp. Hematol., 34, 1462–1470 [DOI] [PubMed] [Google Scholar]
- 168. Cleary H.J, et al. (1999). Allelic loss and promoter hypermethylation of the p15INK4b gene features in mouse radiation-induced lymphoid —but not myeloid —leukaemias. Leukemia, 13, 2049–2052 [DOI] [PubMed] [Google Scholar]
- 169. Koturbash I, et al. (2007). Role of epigenetic effectors in maintenance of the long-term persistent bystander effect in spleen in vivo. Carcinogenesis, 28, 1831–1838 [DOI] [PubMed] [Google Scholar]
- 170. Templin T, et al. (2011). Radiation-induced micro-RNA expression changes in peripheral blood cells of radiotherapy patients. Int. J. Radiat. Oncol. Biol. Phys., 80, 549–557 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Ding N, et al. (2011). Detection of novel human MiRNAs responding to X-ray irradiation. J. Radiat. Res., 52, 425–432 [DOI] [PubMed] [Google Scholar]
- 172. Kumar A, et al. (2011). γ-Radiation induces cellular sensitivity and aberrant methylation in human tumor cell lines. Int. J. Radiat. Biol., 87, 1086–1096 [DOI] [PubMed] [Google Scholar]
- 173. Cui W, et al. (2011). Plasma miRNA as biomarkers for assessment of total-body radiation exposure dosimetry. PLoS One, 6, e22988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Templin T, et al. (2011). Whole mouse blood microRNA as biomarkers for exposure to γ-rays and (56)Fe ion. Int. J. Radiat. Biol., 87, 653–662 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Liu Y, et al. (2008). Aberrant gene promoter methylation in sputum from individuals exposed to smoky coal emissions. Anticancer Res., 28, 2061–2066 [PMC free article] [PubMed] [Google Scholar]
- 176. Liu Y, et al. (2006). Aberrant promoter methylation of p16 and MGMT genes in lung tumors from smoking and never-smoking lung cancer patients. Neoplasia, 8, 46–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Lea J.S, et al. (2004). Aberrant p16 methylation is a biomarker for tobacco exposure in cervical squamous cell carcinogenesis. Am. J. Obstet. Gynecol., 190, 674–679 [DOI] [PubMed] [Google Scholar]
- 178. Huang Y, et al. (2011). Cigarette smoke induces promoter methylation of single-stranded DNA-binding protein 2 in human esophageal squamous cell carcinoma. Int. J. Cancer, 128, 2261–2273 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Lin R.K, et al. (2010). The tobacco-specific carcinogen NNK induces DNA methyltransferase 1 accumulation and tumor suppressor gene hypermethylation in mice and lung cancer patients. J. Clin. Invest., 120, 521–532 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Oka D, et al. (2009). The presence of aberrant DNA methylation in noncancerous esophageal mucosae in association with smoking history: a target for risk diagnosis and prevention of esophageal cancers. Cancer, 115, 3412–3426 [DOI] [PubMed] [Google Scholar]
- 181. Marsit C.J, et al. (2006). Carcinogen exposure and gene promoter hypermethylation in bladder cancer. Carcinogenesis, 27, 112–116 [DOI] [PubMed] [Google Scholar]
- 182. Marsit C.J, et al. (2006). Epigenetic inactivation of the SFRP genes is associated with drinking, smoking and HPV in head and neck squamous cell carcinoma. Int. J. Cancer, 119, 1761–1766 [DOI] [PubMed] [Google Scholar]
- 183. Kikuchi S, et al. (2006). Hypermethylation of the TSLC1/IGSF4 promoter is associated with tobacco smoking and a poor prognosis in primary nonsmall cell lung carcinoma. Cancer, 106, 1751–1758 [DOI] [PubMed] [Google Scholar]
- 184. Anttila S, et al. (2003). Methylation of cytochrome P4501A1 promoter in the lung is associated with tobacco smoking. Cancer Res., 63, 8623–8628 [PubMed] [Google Scholar]
- 185. Kim D.H, et al. (2001). p16(INK4a) and histology-specific methylation of CpG islands by exposure to tobacco smoke in non-small cell lung cancer. Cancer Res., 61, 3419–3424 [PubMed] [Google Scholar]
- 186. Hussain M, et al. (2009). Tobacco smoke induces polycomb-mediated repression of Dickkopf-1 in lung cancer cells. Cancer Res., 69, 3570–3578 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Vaissière T, et al. (2009). Quantitative analysis of DNA methylation profiles in lung cancer identifies aberrant DNA methylation of specific genes and its association with gender and cancer risk factors. Cancer Res., 69, 243–252 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Liu H, et al. (2007). Cigarette smoke induces demethylation of prometastatic oncogene synuclein-gamma in lung cancer cells by downregulation of DNMT3B. Oncogene, 26, 5900–5910 [DOI] [PubMed] [Google Scholar]
- 189. Adenuga D, et al. (2009). Histone deacetylase 2 is phosphorylated, ubiquitinated, and degraded by cigarette smoke. Am. J. Respir. Cell Mol. Biol., 40, 464–473 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Schernhammer E.S, et al. (2010). Dietary folate, alcohol and B vitamins in relation to LINE-1 hypomethylation in colon cancer. Gut., 59, 794–799 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Figueiredo J.C, et al. (2009). Global DNA hypomethylation (LINE-1) in the normal colon and lifestyle characteristics and dietary and genetic factors. Cancer Epidemiol. Biomarkers Prev., 18, 1041–1049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Bönsch D, et al. (2006). Lowered DNA methyltransferase (DNMT-3b) mRNA expression is associated with genomic DNA hypermethylation in patients with chronic alcoholism. J. Neural Transm., 113, 1299–1304 [DOI] [PubMed] [Google Scholar]
- 193. Nan H.M, et al. (2005). Effects of dietary intake and genetic factors on hypermethylation of the hMLH1 gene promoter in gastric cancer. World J. Gastroenterol., 11, 3834–3841 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. D’Addario C, et al. (2011). Ethanol and acetaldehyde exposure induces specific epigenetic modifications in the prodynorphin gene promoter in a human neuroblastoma cell line. FASEB J., 25, 1069–1075 [DOI] [PubMed] [Google Scholar]
- 195. Hicks S.D, et al. (2010). Ethanol-induced methylation of cell cycle genes in neural stem cells. J. Neurochem., 114, 1767–1780 [DOI] [PubMed] [Google Scholar]
- 196. Choi S.W, et al. (1999). Chronic alcohol consumption induces genomic but not p53-specific DNA hypomethylation in rat colon. J. Nutr., 129, 1945–1950 [DOI] [PubMed] [Google Scholar]
- 197. Liu Y, et al. (2009). Alcohol exposure alters DNA methylation profiles in mouse embryos at early neurulation. Epigenetics, 4, 500–511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Hu Y, et al. (2009). Ethanol disrupts chondrification of the neurocranial cartilages in medaka embryos without affecting aldehyde dehydrogenase 1A2 (Aldh1A2) promoter methylation. Comp. Biochem. Physiol. C Toxicol. Pharmacol., 150, 495–502 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Agudelo M, et al. (2011). Effects of alcohol on histone deacetylase 2 (HDAC2) and the neuroprotective role of trichostatin A (TSA). Alcohol. Clin. Exp. Res., 35, 1550–1556 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Zhao Y, et al. (2011). Zinc deprivation mediates alcohol-induced hepatocyte IL-8 analog expression in rodents via an epigenetic mechanism. Am. J. Pathol., 179, 693–702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Avissar M, et al. (2009). MicroRNA expression in head and neck cancer associates with alcohol consumption and survival. Carcinogenesis, 30, 2059–2063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Tang Y, et al. (2008). Effect of alcohol on miR-212 expression in intestinal epithelial cells and its potential role in alcoholic liver disease. Alcohol. Clin. Exp. Res., 32, 355–364 [DOI] [PubMed] [Google Scholar]
- 203. Klaunig J.E, et al. (2000) Epigenetic mechanisms of chemical carcinogenesis. Hum. Exp. Toxicol., 19, 543–555 [DOI] [PubMed] [Google Scholar]
- 204. Melnick R.L, et al. (1996) Implications for risk assessment of suggested nongenotoxic mechanisms of chemical carcinogenesis. Environ. Health Perspect., 104 (suppl. 1), 123–134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. IARC (2012). IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol. 100 IARC, Lyon, France: [Google Scholar]
- 206. Giusti R.M, et al. (1995) Diethylstilbestrol revisited: a review of the long-term health effects. Ann. Intern. Med., 122, 778–788 [DOI] [PubMed] [Google Scholar]
- 207. IARC (1987). IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Supplement 7 IARC, Lyon, France, p. 273 [Google Scholar]
- 208. Newbold R.R, et al. (2006). Adverse effects of the model environmental estrogen diethylstilbestrol are transmitted to subsequent generations. Endocrinology, 147, S11–S17 [DOI] [PubMed] [Google Scholar]
- 209. Newbold R.R, et al. (2002) Characterization of uterine leiomyomas in CD-1 mice following developmental exposure to diethylstilbestrol (DES). Toxicol. Pathol., 30, 611–616 [DOI] [PubMed] [Google Scholar]
- 210. Newbold R.R, et al. (2000). Proliferative lesions and reproductive tract tumors in male descendants of mice exposed developmentally to diethylstilbestrol. Carcinogenesis, 21, 1355–1363 [PubMed] [Google Scholar]
- 211. Lee Y.M, et al. (2011) miRNA-34b as a tumor suppressor in estrogen-dependent growth of breast cancer cells. Breast Cancer Res., 13, R116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Herceg Z, et al. (2011) Epigenetic mechanisms in hepatocellular carcinoma: how environmental factors influence the epigenome. Mutat. Res., 727, 55–61 [DOI] [PubMed] [Google Scholar]
- 213. Arzumanyan A, et al. (2013) Pathogenic mechanisms in HBV- and HCV-associated hepatocellular carcinoma. Nat. Rev. Cancer, 13, 123–135 [DOI] [PubMed] [Google Scholar]
- 214. Calvisi D.F, et al. (2007). Mechanistic and prognostic significance of aberrant methylation in the molecular pathogenesis of human hepatocellular carcinoma. J. Clin. Invest., 117, 2713–2722 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Huang J, et al. (2010). Down-regulated microRNA-152 induces aberrant DNA methylation in hepatitis B virus-related hepatocellular carcinoma by targeting DNA methyltransferase 1. Hepatology, 52, 60–70 [DOI] [PubMed] [Google Scholar]
- 216. Park S.H, et al. (2011) Hepatitis B virus X protein overcomes all-trans retinoic acid-induced cellular senescence by downregulating levels of p16 and p21 via DNA methylation. J. Gen. Virol., 92, 1309–1317 [DOI] [PubMed] [Google Scholar]
- 217. Um T.H, et al. (2011) Aberrant CpG island hypermethylation in dysplastic nodules and early HCC of hepatitis B virus-related human multistep hepatocarcinogenesis. J. Hepatol., 54, 939–947 [DOI] [PubMed] [Google Scholar]
- 218. Horikawa I, et al. (2003). Transcriptional regulation of the telomerase hTERT gene as a target for cellular and viral oncogenic mechanisms. Carcinogenesis, 24, 1167–1176 [DOI] [PubMed] [Google Scholar]
- 219. Cougot D, et al. (2007). The hepatitis B virus X protein functionally interacts with CREB-binding protein/p300 in the regulation of CREB-mediated transcription. J. Biol. Chem., 282, 4277–4287 [DOI] [PubMed] [Google Scholar]
- 220. Fernandez A.F, et al. (2010). Viral epigenomes in human tumorigenesis. Oncogene, 29, 1405–1420 [DOI] [PubMed] [Google Scholar]
- 221. Herceg Z, et al. (2009). HBV protein as a double-barrel shot-gun targets epigenetic landscape in liver cancer. J. Hepatol., 50, 252–255 [DOI] [PubMed] [Google Scholar]
- 222. Pollicino T, et al. (2007). Molecular and functional analysis of occult hepatitis B virus isolates from patients with hepatocellular carcinoma. Hepatology, 45, 277–285 [DOI] [PubMed] [Google Scholar]
- 223. Chiba T, et al. (2012). Inflammation-associated cancer development in digestive organs: mechanisms and roles for genetic and epigenetic modulation. Gastroenterology, 143, 550–563 [DOI] [PubMed] [Google Scholar]
- 224. Uemura N, et al. (2001) Helicobacter pylori infection and the development of gastric cancer. N. Engl. J. Med., 345, 784–789 [DOI] [PubMed] [Google Scholar]
- 225. Katsurano M, et al. (2012). Early-stage formation of an epigenetic field defect in a mouse colitis model, and non-essential roles of T- and B-cells in DNA methylation induction. Oncogene, 31, 342–351 [DOI] [PubMed] [Google Scholar]
- 226. Martin M, et al. (2012). From hepatitis to hepatocellular carcinoma: a proposed model for cross-talk between inflammation and epigenetic mechanisms. Genome Med., 4, 8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Ke Q, et al. (2006). Alterations of histone modifications and transgene silencing by nickel chloride. Carcinogenesis, 27, 1481–1488 [DOI] [PubMed] [Google Scholar]
- 228. Gospodinov A, et al. (2012). Shaping chromatin for repair Mutat. Res., 12, 45–60 [DOI] [PubMed] [Google Scholar]
- 229. Schuster-Böckler B, et al. (2012). Chromatin organization is a major influence on regional mutation rates in human cancer cells. Nature, 488, 504–507 [DOI] [PubMed] [Google Scholar]
- 230. Duhl D.M, et al. (1994). Neomorphic agouti mutations in obese yellow mice. Nat. Genet., 8, 59–65 [DOI] [PubMed] [Google Scholar]
- 231. Wolff G.L, et al. (1998). Maternal epigenetics and methyl supplements affect agouti gene expression in Avy/a mice. FASEB J., 12, 949–957 [PubMed] [Google Scholar]
- 232. Waterland R.A, et al. (2003). Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol. Cell. Biol., 23, 5293–5300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Cooney C.A, et al. (2002). Maternal methyl supplements in mice affect epigenetic variation and DNA methylation of offspring. J. Nutr., 132, 2393S–2400S [DOI] [PubMed] [Google Scholar]
- 234. Jirtle R.L, et al. (2007). Environmental epigenomics and disease susceptibility. Nat. Rev. Genet., 8, 253–262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Rakyan V.K, et al. (2003). Transgenerational inheritance of epigenetic states at the murine Axin(Fu) allele occurs after maternal and paternal transmission. Proc. Natl Acad. Sci. U S A, 100, 2538–2543 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Waterland R.A. (2006) Assessing the effects of high methionine intake on DNA methylation. J. Nutr., 136, 1706S–1710S [DOI] [PubMed] [Google Scholar]
- 237. Druker R, et al. (2004). Complex patterns of transcription at the insertion site of a retrotransposon in the mouse. Nucleic Acids Res., 32, 5800–5808 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Waterland R.A, et al. (2010). Season of conception in rural gambia affects DNA methylation at putative human metastable epialleles. PLoS Genet., 6, e1001252 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Skinner M.K, et al. (2012). Epigenetic transgenerational inheritance of somatic transcriptomes and epigenetic control regions. Genome Biol., 13, R91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Lyko F, et al. (2006) Epigenetic regulation in Drosophila. Curr. Top. Microbiol. Immunol., 310, 23–44 [DOI] [PubMed] [Google Scholar]
- 241. Raddatz G, et al. (2013) Dnmt2-dependent methylomes lack defined DNA methylation patterns. Proc. Natl Acad. Sci. USA, 110, 8627–8631 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Bird A. (2002). DNA methylation patterns and epigenetic memory. Genes Dev., 16, 6–21 [DOI] [PubMed] [Google Scholar]
- 243. Goodman J.I, et al. (2010) What do we need to know prior to thinking about incorporating an epigenetic evaluation into safety assessments? Toxicol. Sci., 116, 375–381 [DOI] [PubMed] [Google Scholar]
- 244. Laird P.W. (2010). Principles and challenges of genomewide DNA methylation analysis. Nat. Rev. Genet., 11, 191–203 [DOI] [PubMed] [Google Scholar]
- 245. Esteller M. (2007). Cancer epigenomics: DNA methylomes and histone-modification maps. Nat. Rev. Genet., 8, 286–298 [DOI] [PubMed] [Google Scholar]
- 246. Umer M, et al. (2013). Deciphering the epigenetic code: an overview of DNA methylation analysis methods Antioxid. Redox Signal., 12, 1972–1986 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Bock C. (2012) Analysing and interpreting DNA methylation data. Nat. Rev. Genet., 13, 705–719 [DOI] [PubMed] [Google Scholar]
- 248. Herceg Z, et al. (2011). New concepts of old epigenetic phenomena and their implications for selecting specific cell populations for epigenomic research. Epigenomics, 3, 383–386 [DOI] [PubMed] [Google Scholar]
- 249. Olivier M, et al. (2012) Upper urinary tract urothelial cancers: where it is A:T. Nat. Rev. Cancer, 12, 503–504 [DOI] [PubMed] [Google Scholar]
- 250. Houseman E.A, et al. (2012). DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics, 13, 86 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Liu Y, et al. (2013). Epigenome-wide association data implicate DNA methylation as an intermediary of genetic risk in rheumatoid arthritis. Nat. Biotechnol., 12 142–147 [DOI] [PMC free article] [PubMed] [Google Scholar]


