IJLSSR, VOLUME 2, ISSUE 4, JULY-2016:385-403

Review Article (Open access)

Effect of Environmental Chemical Exposures on Epigenetics of Diseases:
A Systematic Review

Shovit Ranjan1*, Praveen Kumar Sharma1
1Centre for Life Sciences, Central University of Jharkhand, Brambe, Ranchi, Jharkhand, India

*Address for Correspondence: Shovit Ranjan, Res earch Scholar, Centre for Life Sciences, Central University of Jharkhand,
Brambe, Ranchi, Jharkhand, India

Received: 20 April 2016/Revised: 12 May 2016/Accepted: 14 June 2016

ABSTRACT- Every year more than 13 million deaths worldwide are due to environmental pollutants, and approximately 24% of diseases are caused by environmental exposures that might be averted through preventive measures. Out of all these environmental chemicals, effects of air pollution is responsible for death of 3.3 million people prematurely worldwide - a figure that could double by 2050 if emissions continue to rise at the current rate. Increasing number of evidences has linked environmental pollutants with epigenetic variations, including changes in DNA methylation status, histone modifications and other factors like incorporation of miRNAs, nucleosome remodeling, etc. These entire mechanisms are likely to play important roles in disease aetiology, and their modifications, thus providing further understanding of disease aetiology, as well as biomarkers for these exposures to environmental chemicals and/or prediction of the risk for the disease. In this, we had tried to summarize the different epigenetic alterations related to environmental chemical exposures, and propose the probable mechanisms of action behind such epigenetic changes. We will also focus onopportunities, challenges and further directions for future epidemiology research in environmental epigenomics. Further studies are needed in this regard to solve methodological and practical challenges, including uncertainties about stability over time of epigenomic changes induced by the environment, tissue specificity of epigenetic alterations, validation of laboratory methods, and adaptation of bioinformatic and biostatistical methods to high-throughput epigenomics. Moreover, there are several reports of epigenetic modifications arising from environmental chemical exposures, but most have not been directly linked to disease endpoints.
Key-words- Environmental chemicals, Epigenetics, Disease susceptibility

INTRODUCTION- Being a part of our daily life, chemicals in the day to day use may also cause different diseases through various mechanisms. Environmental pollutants results inapproximately 13 million deaths approximately every year and as much as 24% of the diseases are estimated to be caused by environmental exposures that can be prevented (Pru¨ss-U¨stu¨n Annette, 2006).Out of all these environmental chemicals, effects of air pollution is responsible for death of 3.3 million people prematurely worldwide - a figure that could double by 2050 if emissions continue to rise at the current rate (Lelieveld et al., 2015).
Growing evidence suggests that environmental pollutants may cause diseases via epigenetic mechanism-regulated gene expression changes (Tang et al., 2007; Bezeket al., 2008). Continuous exposure to many chemicals, including through air, water, food or other media and products resulting in various diseases and health impacts are well assessed, however very little is known about the mechanism at the epigenetic level. This review has tried to summarize the effect of different environmental chemical exposures on epigenetics of various diseases studied till now (Table 1).

Epigenetics-Linking Factor between Environment and different diseases- Epigenetics defined as heritable changes in gene function occuring without a change in the nucleotide sequence (Bird, 2007). These changes in phenotypic traits occur due to variety of mechanisms (Fradin and Bougneres, 2011). An Epigenetic factor that regulates gene expression mostly includes DNA methylations, histone modifications, and expression of microRNAs (miRNAs) (Reik et al., 2001; Grewal and Moazed, 2003). An epigenetic mechanism that modifies chromatin structure can be classified into four main categories: DNA methylation, covalent histone modifications, and non-covalent mechanisms like incorporation of histone variants and nucleosome remodeling and non-coding RNAs including microRNAs (miRNAs).

Epigenetic Changes due to environmental chemical exposures- Changes in these epigenetic factors have been shown to be induced by the exposure to various environmental chemicals linked with different diseases (Baccarelli et al., 2009; Heightman et al., 2011; Wright, 2011).Entire list of such epigenetic changes as described by Hou, Zhang, Wang and Baccarelli due to different environmental factors like pollution, chemicals, pesticides, etc are enlisted in Table 1. Various epigenetic mechanisms responsible for it are described below as follows-

DNA Methylation- Out of all, DNA methylation is the mostthoroughly studied epigenetic modification in mammals, playing an important role in regulating gene expression and chromatin architecture, in association with histone modifications and other chromatin associated proteins. DNA methylation mainly occurs by the covalent modification of cytosine residues in CpG dinucleotides in mammals. In human genome, CpG dinucleotides are not evenly distributed across the human genome but are instead concentrated in ‘CpG islands’ and regions of large repetitive sequences (e.g. centromeric repeats, retrotransposon elements, rDNA etc.) (Bird, 2002; Takai et al., 2002).
During development and in differentiated tissues, most of the CpG sites in the genome are methylated, but the mostof the CpG islands usually remain unmethylated also (Suzuki et al., 2008). However, some CpG island promoters get methylated during development, resulting in long-term transcriptional silencing (Bird, 2002). DNA methylation uses various mechanisms to heritably silence genes and non-coding genomic regions. DNA methylation can lead to gene silencing by either preventing or promoting the recruitment of regulatory proteins to DNA (Prendergastet al., 1991; Wattet al., 1988) or can also mediate gene repression through interactions with histone deacetylases (HDACs) (Jones et al., 1998; Nan et al., 1998).
Recent studies have suggested that DNA methylation is also important for the regulation of non- CpG island promoters (Futscheret al., 2002; Hattoriet al., 2004). In order to fully understand the global role of DNA methylation in normal tissue,it is essential to elucidate the role of non-CpG island methylation, as CpG islands has been found to occupy only approximately of 60% of human gene promoters(Wang et al., 2004).

Covalent Histone Modifications- Histone proteins consist of the nucleosome core, havinga globular C-terminal domain and N-terminal tail (Luger et al., 1997). The N-terminal tails of histones can undergo a variety of posttranslational covalent modifications like methylation, acetylation, ubiquitylation, sumoylation and phosphorylation on specific amino acid residues, resulting in the regulation of key cellular processes such as transcription, replication and repair (Kouzarides, 2007). These modifications are proposed to store the epigenetic memory inside a cell in the form of a ‘histone code’ that determines the structure and activity of different chromatin regions (Jenuweinet al., 2001). Histone modifications take place by either changing the chromatin accessibility or by recruitment of non-histone effector proteins. The mechanism of inheritance of the histone code, however, is still not fully understood.

Nucleosome Remodelling and Histone Variants- Non-covalent mechanism of nucleosome remodeling and presence of specialized histone variants, sometimes also plays an important role in regulation of chromatin structure and gene activity. Nucleosomes regulate gene expression by altering the accessibility of regulatory DNA sequences to transcription factorsin addition to its function as DNA packaging within a cell (Jiang et al., 2009). Nucleosome free regions (NFRs) present at the 5’ and 3’ends of genes provide the sites for assembly and disassembly of the transcription machinery (Yuan et al., 2005). The nucleosome loss directly upstream of the transcription start site is strongly correlated with gene activation (Shivaswamy et al., 2008; Lin et al., 2007). Moreover, the presence of an NFR at gene promoters with basal level of transcription is related with the ability for rapid activation upon stimulation (Gal-Yam et al., 2006). In contrast, shutting off of the transcription start site within the NFR by a nucleosome is associated with gene repression (Schones et al., 2008). NFR modulation is achieved by ATP-dependent chromatin-remodeling complexes, which modifies the accessibility of DNA regulatory sitesthrough both sliding and ejection of nucleosomes (Smith et al., 2005). The interaction between nucleosome remodeling machinery, DNA methylation and histone modifications plays a vital role in establishing global gene expression patterns and chromatin design (Harikrishnan et al., 2005; Wysocka et al., 2006).

Non-coding RNA like miRNAs- miRNAs are small, approximately 22 nucleotides, non-coding RNAs that regulate gene expression through posttranscriptional silencing of target genes. Sequence-specific base pairing of miRNAs with 3’untranslated regions of target mRNA within the RNA-induced silencing complex results in degradation of target messenger RNA or inhibition of translation (He et al., 2004). miRNAs are expressed in a tissue-specific manner and control a wide array of biological processes including cell proliferation, apoptosis and differentiation. The list of miRNAs identified in the human genome and their potential target genes is growing rapidly, demonstrating their extensive role in maintaining global gene expression patterns (Zhang et al., 2007).
Like normal genes, the expression of miRNAs can be regulated by epigenetic mechanisms (Saito et al., 2006). In addition, miRNAs can also modulate epigenetic regulatory mechanisms inside a cell by targeting enzymes responsible for DNA methylation (DNMT3A and DNMT3B) and histone modifications (EZH2) (Fabbriet al., 2007; Friedman et al., 2009). Such interaction among the various components of the epigenetic machinery re-emphasizes the integrated nature of epigenetic mechanisms involved in the maintenance of global gene expression patterns.

Table 1: Effect of Environmental Chemicals on epigenetic changes of various diseases

Environmental Chemicals Epigenetic Changes Details of study Diseases studied References
Air Pollution DNA methylation-
Global hypomethylation
Human PBL(In vivo) Various cancers and
Schizophrenia
Baccarelli et al.,2009;
Smith et al.,2007;
Roman-Gomez et al.,2006;
Deng et al.,2006;
Brothman et al.,2005;
Shimabukuro et al.,2007
iNOS hypomethylation Human PBL(In vivo) Lung cancer Tarantini et al.,2009;
Pereira et al.,2007
Global hypermethylation C57BL/CBA mice
sperm(In vivo)
Colorectal cancer
renal cell carcinoma,
acute lymphoblastic
leukaemia and bladder
urothelial cell carcinoma
Yauk et al.,2008;
Cheetham et al.,2008;
Alemayehu et al.
Norrie et al.,2002;
Minardi et al.,2009;
Schafer et al.,2010;
Owen et al.,2010
Hypermethylation of IFNg
and hypomethylation of IL4
CD4+T-Lymphocytes(In vivo) Asthma Liu et al.,2008
Histone modification -
Increased H3K4
dimethylation and H3K9
acetylation
Human PBL(In vivo) Diabetic nephropathy Cantone et al.,2011; Sayyed et al.,2010
Global hypomethylation
(Alu, LINE-1)
Human buffy coat(In vivo) Various cancers and
schizophrenia
Klein et al.,2002;
Smith et al.,2007;
Roman-Gomez et al.,2006;
Deng et al.,2006;
Brothman et al.,2005;
Shimabukuro et al.,2007
miRNAs-
Increased miR-222
Human PBL(In vivo) Various cancers Klein et al.,2002;
Felli et al.,2005;
le Sage et al.,2007;
Garofalo et al.,2009
Increased miR-21 Human PBL(In vivo) Various cancers Klein et al.,2002;
Connolly etal.,2008;
Chan et al.,2005;
Iorio et al.,2005;
Frankel et al.,2008;
Zhu et al.,2007;
Schetter et al.,2008;
Bloomston et al.,2007;
Meng et al.,2007
Aluminium miRNAs-
Increased miR-146a
HN cells(In vitro) AD, cardiac hypertrophy
and various cancers
Pogue et al.,2009;
Lukiw et al.,2008;
Pogue et al., 2009;
Cheng et al.,2007;
Volinia et al.,2006;
Taganov et al.,2006;
Bhaumik et al.,2008;
Shen et al.,2008;
Calin et al.,2005;
Xu et al.,2008;
Yanaihara et al.,2006;
Kozaki et al.,2008
Increased miR-9, miR-128,
miR-125b
HN cells(In vitro) AD, neurodegeneration
and various cancers
Lukiw et al.,2007;
Saba et al.,2008;
Roehle et al.,2008;
Wang et al.,2008;
Tan et al.,2010;
Veerla et al.,2009
Arsenic DNA methylation-
Global hypomethylation
Human HaCaT keratinocytes,
human prostate
epithelial cell line
RWPE-1, TRL 1215 rat
liver epithelial cell line,
V79-Cl3 Chinese hamster
cells(In vitro)
Various cancers and
schizophrenia
Reichard et al.,2007;
Benbrahim-Tallaa et al.,2005;
Coppin et al.,2008;
Zhao et al.,1997;
Sciandrello et al.,2004;
Smith et al.,2007;
Roman-Gomez et al.,2006;
Deng et al.,2006;
Brothman et al.,2005;
Shimabukuro et al.,2007
129/SvJ mice,84 fisher
344 Rat,86 homozygous
Tg.AC mice,87
goldfish,232 human
PBL233(In vivo)
Various cancers and
schizophrenia
Chen et al.,2004;
Uthus et al.,2005;
Xie et al.,2004;
Smith et al.,2007;
Roman-Gomez et al.,2006;
Deng et al.,2006;
Brothman et al.,2005;
Shimabukuro et al.,2007
Global hypomethylation
and c-Ha-ras
hypomethylation
C57BL/6J mice(In vivo) Various cancers and
schizophrenia
Okoji et al., 2002;
Smith et al., 2007;
Roman-Gomez et al.,2006;
Deng et al.,2006;
Brothman et al.,2005;
Shimabukuro et al.,2007
Global hypermethylation Human PBL(In vivo) Colorectal cancer, renal
cell carcinoma, acute
lymphoblastic leukaemia
and bladder urothelial cell
carcinoma
Majumdar et al.,2010;
Pilsner et al.,2007;
Cheetham et al.,2008;
Alemayehu et al.,2008;
Norrie et al.,2002;
Minardi et al.,2009;
Schafer et al.,2010;
Owen et al.,2010;
DAPK hypermethylation SV-HUC-1 cells(In vitro) Various cancers Chai et al.,2007;
Qian et al.,2010;
Laytragoon-Lewin et al.,2010;
Paluszczak et al.,2011;
Hafner et al.,2011;
Li et al.,2011;
Ben Ayed-Guerfali et al.,2011;
Sugita et al.,2011;
Zhang et al.,2011;
Hu et al.,2010;
Van der Auwera et al.,2010;
Zhang et al.,2011;
Peng et al.,2010
P16 hypermethylation Human myeloma cell line U266(In vitro) Various cancers Lewin et al.,2010;
Hu et al.,2010;
Zhang et al.,2011;
Malhotra et al.,2010;
Poetsch et al.,2011;
Lin et al.,2012;
Wang et al.,2011;
Zainuddin et al.,2011;
Shaw et al.,2010
DBC1, FAM83A,
ZSCAN12 and C1QTNF6
Human UROtsa cells(In vitro) Bladder cancer, breast
cancer and malignant
Jensenet al.,2008;
Serizawa et al.,2011;
Hill et al.,2010;
hypermethylation lymphoproliferative
neoplasms
Gronbaek et al.,2008
P53 hypermethylation Human lung adenocarcinoma
A549 cells(In vitro)
Breast cancer and
hepatoblastoma
Mass et al.,1997;
Radpour et al.,2010;
Hanafusa et al.,2005
C-myc hypomethylation TRL 1215 rat liver
epithelial cells(In vitro)
cancer, liver cancer,
kidney cancer and bladder
cancer
Chen et al.,2001;
Pereira et al.,2001;
Luo et al.,2010;
Fang et al.,1996;
Tsujiuchi et al.,1999;
Shen et al.,1997;
Del et al.,1989
C-myc and c-Ha-ras
hypomethylation
Syrian hamster embryo
cells (In vitro)
Gastric cancer, colon cancer,
liver cancer, kidney
cancer and bladder cancer
Takahashi et al.,2002;
Pereira et al.,2001;
Luo et al.,2010;
Fang et al.,1996;
Tsujiuchi et al.,1999;
Shen et al.,1997;
Del et al.,1989
P16 and RASSF1
hypermethylation
A/J mice (In vivo) Various cancers Cui et al.,2006;
Laytragoon-Lewin et al.,2010;
Hu et al.,2010;
Zhang et al.,2011;
Malhotra et al.,2010;
Poetsch et al.,2011;
Lin et al.,2012;
Wang et al.,2011;
Zainuddin et al.,2011;
Shaw et al.,2010;
Rabiau et al.,2009;
Buckingham et al.,2010
Global hypomethylation
and ER-alpha
hypomethylation
C3H mice (In vivo) Various cancersand
schizophrenia
Waalkes et al.,2004;
Smith et al.,2007;
Roman-Gomez et al.,2006;
Deng et al.,2006;
Brothman et al.,2005;
Shimabukuro et al.,200
P53 and P16
hypermethylation
Human PBL (In vivo) Various cancers Chanda et al.,2006;
Laytragoon- Lewin et al.,2010;
Hu et al.,2010;
Zhang et al.,2011;
Malhotra et al.,2010;
Poetsch et al.,2011;
Lin et al.,2012;
Wang et al.,2011;
Zainuddin et al.,2011;
Shaw et al.,2010;
Radpour et al.,2010;
Hanafusa et al.,2005
DAPK hypermethylation Human bladder, kidney
and ureter (In vivo)
Various cancers Chen et al.,2007;
Qian et al.,2010;
Laytragoon-Lewin et al.,2010;
Paluszczak et al.,2011;
Hafner et al.,2011;
Li et al.,2011;
Ben Ayed-Guerfali et al.,2011;
Sugita et al.,2011;
Zhang et al.,2011;
Hu et al.,2010;
Van der Auwera et al.,2010;
Zhang et al.,2011;
Peng et al.,2010
RASSF1A and PRSS3
hypermethylation
Human bladder (In vivo) Lung cancer and prostate
cancer
Marsit et al.,2006;
Rabiau et al.,2009;
Buckingham et al.,2010
P16 hypermethylation Human PBL (In vivo) Various cancers Zhang et al.,2007;
Laytragoon Lewin et al.,2010;
Hu et al.,2010;
Zhang et al.,2011;
Malhotra et al.,2010;
Poetsch et al.,2011;
Lin et al.,2012;
Wang et al.,2011;
Zainuddin et al.,2011;
Shaw et al.,2010
P53 hypermethylation Human basal cell
carcinoma(In vivo)
Breast cancer and
hepatoblastoma
Boonchai et al.,2000;
Radpour et al.,2010;
Hanafusa et al.,2005
Both hypomethylation and
hypermethylation of VHL
Human kidney cells (In vitro) Renal cell carcinoma Zhong et al.,2001
Histone modification-
Decreased H3 acetylation
UROtsa and URO-ASSC cells(In vitro) Renal cell carcinomas Jensen et al.,2008;
Kanao et al.,2008
Decreased H4K16
acetylation
UROtsa cells (In vitro) Bladder cancer Jo et al.,2009
Increased H3K14
acetylation
Increased H3S10
phosphorylation
NB4 cells (In vitro) Diabetic nephropathy Li et al.,2002;
Sayyed et al.,2010
Increased H3
phosphorylation
WI-38 human diploid
fibroblast cells(In vitro)
Diabetic nephropathy Li et al.,2003;
Sayyed et al.,2010
Increased H3K9
acetylation
HepG2 hepatocarcinoma
cells(In vitro)
Diabetic nephropathy Ramirez et al.,2008;
Sayyed et al.,2010
Decreased H3, H4, H2a,
H2b acetylation
Decreased H3 and H4
methylation
Increased H2b methylation
Drosophila melanogaster tissue culture cell line KC161(In vitro) Heart disease and traumatic
brain injury
Arrigo et al.,1983;
Gaikwad et al.,2010;
Gao et al.,2006
Increased H3K36
trimethylation
Decreased H3K36
dimethylation
Increased H3K4
dimethylation
Human lung carcinoma A549 cells(In vitro) Diabetic nephropathy,
multiple myeloma and
prostate cancer
Zhou et al.,2008;
Sayyed et al.,2010;
Zhao et al.,2010;
Seligson et al.,2009
Increased H3K9
dimethylation
Decreased H3K27
trimethylation
Increased H3K4
trimethylation
Human lung carcinoma A549 cells(In vitro) Prostate cancer, kidney
cancer, lung cancer, HCC
and AML
Zhou et al.,2008;
Seligson et al.,2009;
Arita et al.,2009;
Chen et al.,2010;
Yao et al.,2010;
Paul et al.,2010
Increased H2AX
phosphorylation
Decreased H3K18
acetylation
Decreased H3R17
methylation
RPMI7951 melanoma
cells(In vitro)
1470.2 cell line derived
from the mouse a denocarcinoma parent
line(In vitro)
Ataxia telangiectasia
Prostate cancerand colon
cancer
Zykova et al.,2006;
Porcedda et al.,2008
Barr et al.,2009;
Seligson et al.,2009;
Ashktorab et al.,2009

miRNAs-
Increased miR-222, Decreased miR-210
TK6 cell line(In vitro) Various cancersand AD Marsit et al.,2006;
Felli et al.,2005;
le Sage et al.,2007;
Garofalo et al.,2009;
Mi et al.,2007;
Saumet et al.,2009;
Hebert et al.,2008
Decreased miR-19a T24 cell line(In vitro) Various cancers Cao et al.,2011;
Takakura et al.,2008;
Calin et al.,2004;
Arndt et al.,2009;
Bandres et al.,2006;
Malzkorn et al.,2010;
Hebert et al.,2007;
Budhu et al.,2008;
Connolly et al.,2008;
Hayashita et al.,2005
Benzene DNA methylation-
Global hypomethylation
(Alu, LINE-1)
Human PBL (In vivo) Various cancers and
schizophrenia
Baccarelli et al.,2009;
Smith et al.,2007;
Roman-Gomez et al.,2006;
Deng et al.,2006;
Brothman et al.,2005;
Shimabukuro et al.,2007
P15 hypermethylation and
melanoma antigen-1
(MAGE-1)
hypomethylation
Human PBL (In vivo) Psoriasis and various
cancers
Kim et al.,2007;
Bassil et al.,2007;
Koutros et al.,2010;
Waggoner et al.,2011;
Bollati et al.,2007;
Zhang et al.,2009;
Furonaka et al.,2004;
Lindberg et al.,2008;
Kim et al.,2009;
Shimamoto et al.2005;
Chen et al.,2002;
Gallardo et al.,2004;
El-Shakankiry et al.,2006;
Matsuno et al.,2005;
Wemmert et al.,2009;
Berg et al.,2007;
Wong et al.,2003
Global DNA hypomethylation Human lymphoblastoid
cell line TK6(In vitro)
Various cancers and
schizophrenia
Ji et al.,2010;
Smith et al.,2007;
Roman-Gomez et al.,2006;
Deng et al.,2006;
Brothman et al.,2005;
Shimabukuro et al.,2007
Hypermethylation of poly
(ADP-ribose) polymerases-
1 (PARP-1)
Lymphoblastoid cell line
F32(In vitro)
Various cancers Gao et al.,2010
Bisphenol A DNA methylation-
Hypomethylation of the
Agouti gene and CabpIAP
Mouse embryo(In vivo) Mice with
hypomethylation of the
Agouti gene are obese,
diabetic and exhibit
increased cancer rates
Dolinoy et al.,2007;
Morgan et al.,2010;
Xiang et al.,2010
Hypomethylation of the
homeobox gene Hoxa10
CD-1 mice (In vivo) Not applicable Bromer et al.,2010;
Hypermethylation of
LAMP3.
Breast epithelial cells
(In vitro)
Breast cancer Weng et al.,2010;
miRNAs-
Increased miR-146a
3A placental cells
(In vitro)
Cardiac hypertrophy,
AD and various cancers
Whiting et al.,2010;
Lukiw et al.,2008;
Pogue et al.,2009;
Cheng et al.,2007;
Volinia et al.,2006;
Taganov et al.,2006;
Bhaumik et al.,2008;
Shen et al.,2008;
Calin et al.,2005;
Xu et al.,2008;
Yanaihara et al.,2006;
Kozaki et al.,2008;
Cadmium DNA methylation-
Global DNA
hypomethylation
K562 cell(In vitro) Colorectal cancer, renal
cell carcinoma, acute
lymphoblastic
leukaemia,bladder
urothelial cell carcinoma
Huang et al.,2008;
Cheetham et al.,2008;
Alemayehu et al.,2008;
Norrie et al.,2002;
Minardi et al.,2009;
Schafer et al.,2010;
Owen et al.,2010
Initially induces DNA
hypomethylation,
prolonged exposure results
in DNA hypermethylation
TRL1215 rat liver cells
(In vitro)
Not applicable Takiguchi et al.,2003;
miRNAs-
Decreased miR-146a
Human PBL(In vivo) Various cancers Bollati et al.,2010;
Gramantieri et al.,2007;
Jazdzewski et al.,2008;
Lin et al.,2008
Chromium DNA methylation-
P16 and hMLH1
hypermethylation
Human lung(In vivo) Various cancers Kondo et al.,2006;
Takahashi et al.,2005;
Laytragoon-Lewin et al.,2010;
Hu et al.,2010;
Zhang et al.,2011;
Malhotra et al.,2010;
Poetsch et al.,2011;
Lin et al.,2012;
Wang et al.,2011;
Zainuddin et al.,2011;
Shaw et al.,2010;
Gonzalez- Ramirez et al.,2011;
Vasavi et al.,2010;
Ling et al.,2010
Gpt hyperme,thylation G12 cell line (In vitro) Not applicable Klein et al.,2002
Histone modification-
Decreased H3S-10
Phosphorylation
Human lung carcinoma
A549 cells (In vitro)
Type 2 diabetes, heart
disease and traumatic
brain injury
Arita et al.,2009; Sayyed et al.,2010;
Gaikwad et al.,2010;
Gao et al.,2006
Decreased H3K4
trimethylation
Decreased H3 and H4
acetylation
Increased Dimethylation
and trimethylation of
H3K9 and H3K4
Decreased
H3K27trimethylation and
H3R2 dimethylation
DES miRNAs-
Decreased miR-9-3
Breast epithelial cells (In vitro) Breast cancer Hsu et al.,2009
Dioxin DNA methylation- Igf2 hypomethylation Rat liver (In vivo) Russell–Silver syndrome
and various cancers
Wang et al.,2007;
Gucev et al.,2009;
Zeschnigk et al.,2008;
Chopra et al.,2010;
Dammann et al.,2010;
Baba et al.,2010;
Li et al.,2009;
Cui et al.,2002;
Ito et al.,2008
Alterations in DNA methylation
at multiple genomic
regions
Splenocyte of mice (In vivo) Not applicable McClure et al.,2011
Drinking Water DNA methylation-
Global hypomethylation
c-myc hypomethylation
Mice liver (In vivo) Gastric cancer, colon
cancer, liver cancer,
kidney cancer and bladder cancer
al.,2000; Luoet al.,2010;
Fang et al.,1996;
Tsujiuchi et al.,1999;
Shen et al.,1997;
Del et al.,1989
Lead DNA methylation-
Global hypomethylation
Human PBL, newborn
umbilical cord blood
samples(In vivo)
Various cancersand
schizophrenia
Wright et al.,2010;
Pilsner et al.,2009;
Smith et al.,2007;
Roman-Gomez et al.,2006;
Deng et al.,2006;
Brothman et al.,2005;
Shimabukuro et al.,2007
Mercury DNA methylation-
Global hypomethylation
Brain tissues in polar
bear(In vivo)
Neurological disorders
and various cancer
Pilsner et al.,2010;
Mill et al.,2008;
Wang et al.,2008;
Esteller et al.,2008
Rnd2 hypermethylation Mouse embryonic stem
cells(In vitro)
Neuronal migration defect Arai et al.,2011;
Heng et al.,2008
Nickel DNA methylation-
ATF-1, HIF-1, gpt and Rb
hypermethylation
G12 cell line(In vitro) Various cancers Lee et al.,1995;
Chim et al.,2003;
Stirzaker et al.,1997;
Chen et al.,2004;
Zhao et al.,2010;
Zhao et al.,2003;
Li et al.,1998
P16 hypermethylation Mouse histiocytomas(In vivo) Various cancers Govindarajan et al.,2002;
Laytragoon-Lewin et al.,2010;
Hu et al.,2010;
Zhang et al.,2011;
Malhotra et al.,2010;
Poetsch et al.,2011;
Lin et al.,2012;
Wang et al.,2011;
Zainuddin et al.,2011;
Shaw et al.,2010
Histone modification-
Increased H3K9
methylation
Decreased Acetylation at
all four core histones
Human lung carcinoma
A549 cells(In vitro)
Heart disease and
traumatic brain injury
Chen et al.,2006;
Ke et al.,2006;
Gaikwad et al.,2010;
Gao et al.,2006
dimethylation
Increased H2a, H2b
ubiquitylation
Decreased H3K4
methylation
Decreased H3K4
acetylation
Decreased H2a, H2b, H3,
H4 acetylation
Human lung carcinoma
A549 cells,G12 cells,
1HAEo- cell line, human (HAE) and rat (NRK)
cells, Chinese hamster
cell line (In vitro)
Lung cancer, heart
disease, chronic
glomerular disease and
traumatic brain injury
Lee et al.,1995;
Karaczyn et al.,2005;
Karaczyn et al.,2006;
Broday et al.,2000;
Chen et al.,2006;
Ke et al.,2006;
Klein et al.,1997;
Yan et al.,2003;
Arita et al.,2009;
Gaikwad et al.,2010;
Gao et al.,2006;
Chen et al.,2010;
Lefevre et al.,2010
DecreasedH4K5, H4K8,
H4K12, H4K16 acetylation
Human lung carcinoma
A549 cells (In vivo)
Ataxia telangiectasia Broday et al.,2000;
Kumar et al.,2011
DecreasedH2A, H2B, H3,
H4 acetylation (especially
in H2BK12 and H2BK20)
Human airway epithelial
1HAEo(HAE) cell line
(In vitro)
Heart disease and
traumatic brain injury
Golebiowski et al.,2005;
Gaikwad et al.,2010;
Gao et al.,2006;
Increased H3 phosphorylation Human lung carcinoma
A549 cells (In vitro)
Diabetic nephropathy Ke et al., 2008;
Sayyed et al., 2010
Pesticides DNA methylation-
P53 hypermethylation
Human lung adenocarcinoma
A549 cells (In vitro)
Breast cancer and
hepatoblastoma
Mass et al.,1997;
Radpour et al.,2010;
Hanafusa et al.,2005
Alter DNA methylation in
the germ line
Rat testis (In vivo) Potential effects in the
offspring
Anway et al.,2005;
Guerrero-Bosagn et al.,2010;
Anway et al.,2006
Hypomethylation of c-jun
and c-myc
Mouse liver (In vivo) Gastric cancer, colon
cancer, liver cancer, kidney
cancer and bladder
Tao et al.,2000;
Pereira et al.,2001;
Luo et al.,2010;
Fang et al.,1996;
Tsujiuchi et al.,1999;
Shen et al.,1997
Del et al.,1989
Global hypomethylation
(Alu)
Human PBL(In vivo) Various cancersand
schizophrenia
Rusiecki et al.,2008;
Kim et al.,2010;
Smith et al.,2007;
Roman-Gomez et al.,2006;
Deng et al.,2006;
Brothman et al.,2005;
Shimabukuro et al.,2007
Both hypomethylation and
hypermethylation of VHL
Human kidney cells
(In vitro)
Renal cell carcinoma Zhong et al.,2001
Histone modification-
Increased Ac of H3 and H4
Immortalized rat
mesencephalic/ dopaminergic
cells (N27 cells)(In vitro and in vivo)
Parkinson’s disease Song et al.,2010
RDX miRNAs-
Increased let-7, miR-15,
miR-16, miR-26, miR-181
Decreased miR-10b
Mouse brain and liver
(In vivo)
Various cancers Zhang et al.,2009;
Calin et al.,2005;
Calin et al.,2002;
Cimmino et al.,2005;
Ambs et al.,2008;
Roccaro et al.,2009;
Johnson et al.,2005;
Lee et al.,2007;
Yanaihara et al.,2006;
Sampson et al.,2007
Increased miR-206,
miR-30, miR-195
Mouse brain and liver
(In vivo)
Various cancers Zhang et al.,2009;
Iorio et al.,2005;
Adams et al.,2007;
Roccaro et al.,2009;
Dixon- McIver et al.,2008;
van Rooij et al.,2006;
Sayed et al.,2007
PBL: Peripheral blood leucocytes; HCC: Hepatocellular carcinoma; AML: Acute myeloid leukaemia; AD: Alzheimer’s disease; HN cells:
Human neural cells; RDX: Hexahydro-1,3,5-trinitro-1,3,5-triazine; DES: Diethylstilbestrol.


Suitable study designs, approaches, challenges and opportunities for Environmental Epigenomics Studies- The rapid growth of environmental epigenetics field in the past several years has led the investigators to face different difficulties and challenges as well. Few studies had produced uneven results on same environmental chemicals that may be because of several factors. The fact that these tissue specific epigenetic alterations (Minard et al., 2009) is likely to be acceptable because same environmental chemical might produce different epigenetic changes in different tissues, and even it can change within the same tissue on different cell types. Difference in study design, laboratory methods and small sample size may also be major causes for these inconsistencies in epigenetic changes. Replication of results and identification of the sources of variability across studies is one of the major challenges for epigenetic investigations. There relationship between a disease and an epigenetic marker can be determined by an effect of disease on the epigenetic patterns, instead of vice versa (Reltonet al., 2010), since epigenetic markers change over time. The epigenetic alterations that were found to be induced by or associated with environmental pollutants were also found in various diseases. Earlier prospective epidemiological studies might be helpful for mapping epigenomic changes in response to specific chemicals. Methods of collection and processing can modify the cell types stored, thus potentially having its effect on epigenetic marks. In addition to this, high through put methods providing good quality data on DNA methylation, histone modifications and miRNA expression are gradually used these days in human investigations. The share of the effects of any particular environmental exposure that can mediate through epigenetic mechanisms is still undetermined, though epigenetic mechanisms are ideal molecular intermediates of environmental effects. Statistical approaches, including well-designed prospective studies and advanced statistical methods are urgently needed for causal inference in this regard. The epidemiological causal reasoning in epigenomics should include careful consideration of knowledge, data, methods and techniques from several disciplines similar to genomic studies (Geneletti et al., 2011).

Epigenomics: Can it be used for prevention of various diseases- One of the main objectives behind these epidemiology investigations is to look for future preventive interventions. Various clinical and preclinical studies has already showed that most of the epigenetic changes are reversible, which offers novel insights to develop new preventive and therapeutic strategies in this field that can make use of molecules that alter the activities of epigenetic enzymes, such as DNA Methyl Transferases (DNMTs) and Histone Deacetylases (HDACs). Drugs have already been designed and developed in this regard that produce functional effects like histone acetylation and DNA hypomethylation that can be used to restore the normal gene transcription. Future epidemiology studies and epigenomic research to evaluate the effects of environmental exposures on the epigenome may provide information for developing preventive strategies, including exposure reduction, along with pharmacological, dietary or lifestyle interventions as well.

REFERENCES:

  1. Adams BD, Furneaux H, White BA. The microribonucleic acid (miRNA) miR-206 targets the human estrogen receptor-alpha (ER alpha) and represses ERalpha messenger RNA and protein expression in breast cancer cell lines. Mol Endocrinol, 2007; 21:1132–47.
  2. Alavanja MC, Ward MH, Reynolds P. Carcinogenicity of agricultural pesticides in adults and children. J Agromedicine, 2007; 12:39–56.
  3. Alemayehu A, Sebova K, Fridrichova I. Redundant DNA methylation in colorectal cancers of Lynch-syndrome patients. Genes Chrom Cancer, 2008; 47:906–14.
  4. Ambs S, Prueitt RL, Yi M et al. Genomic profiling of microRNA and messenger RNA reveals deregulated microRNA expression in prostate cancer. Cancer Res, 2008; 68:6162–70.
  5. Anway MD, Cupp AS, Uzumcu M, Skinner MK. Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science, 2005; 308:1466–69.
  6. Anway MD, Skinner MK. Epigenetic transgenerational actions of endocrine disruptors. Endocrinology 2006; 147 (6 Suppl):S43–49.
  7. Arai Y, Ohgane J, Yagi S et al. Epigenetic Assessment of environmental chemicals detected in maternal peripheral and cord blood samples. J Reprod Dev, 2011; 57:507–17.
  8. Arita A, Costa M. Epigenetics in metal carcinogenesis: Nickel, Arsenic, Chromium and Cadmium. Metallomics, 2009; 1:222–28.
  9. Arndt GM, Dossey L, Cullen LM et al. Characterization of global microRNA expression reveals oncogenic potential of miR-145 in metastatic colorectal cancer. BMC Cancer, 2009; 9:374–90.
  10. Arrigo AP. Acetylation and methylation patterns of core histones are modified after heat or arsenite treatment of Drosophila tissue culture cells. Nucleic Acids Res 1983; 11: 1389–404.
  11. Ashktorab H, Belgrave K, Hosseinkhah F et al. Global histone H4 acetylation and HDAC2 expression in colon adenoma and carcinoma. Digestive Dis Sci 2009; 54: 2109–17.
  12. Avissar-Whiting M, Veiga KR, Uhl KM et al. Bisphenol A exposure leads to specific microRNA alterations in placental cells. Reprod Toxicol, 2010; 29:401–06.
  13. Baba Y, Nosho K, Shima K et al. Hypomethylation of the IGF2 DMR in colorectal tumors, detected by bisulfite pyrosequencing, is associated with poor prognosis. Gastroenterology, 2010; 139:1855–64.
  14. Baccarelli A, Bollati V. Epigenetics and environmental chemicals. Curr Opin Pediatr, 2009; 21:243–51.
  15. Bagnyukova TV, Luzhna LI, Pogribny IP, Lushchak VI. Oxidative stress and antioxidant defenses in goldfish liver in response to short-term exposure to arsenite. Environ Mol Mutagen, 2007; 48:658–65.
  16. Bandres E, Cubedo E, Agirre X et al. Identification by Real-time PCR of 13 mature microRNAs differentially expressed in colorectal cancer and non-tumoral tissues. Mol Cancer, 2006; 5:29–38.
  17. Barr FD, Krohmer LJ, Hamilton JW, Sheldon LA. Disruption of histone modification and CARM1 recruitment by arsenic represses transcription at glucocorticoid receptor-regulated promoters. PLoS One 2009; 4:e6766.
  18. Bassil KL, Vakil C, Sanborn M, Cole DC, Kaur JS, Kerr KJ. Cancer health effects of pesticides: systematic review. Can Fam Physician, 2007; 53:1704–11.
  19. Ben Ayed-Guerfali D, Benhaj K, Khabir A et al. Hypermethylation of tumor-related genes in Tunisian patients with gastric carcinoma: Clinical and biological significance. J Surg Oncol, 2011;103:687–94.
  20. Benbrahim-Tallaa L, Waterland RA, Styblo M, Achanzar WE, Webber MM, Waalkes MP. Molecular events associated with arsenic-induced malignant transformation of human prostatic epithelial cells: aberrant genomic DNA methylation and K-ras oncogene activation. Toxicol Appl Pharmacol, 2005; 206:288–98.
  21. Berg T, Guo Y, Abdelkarim M, Fliegauf M, Lubbert M. Reversal of p15/INK4b hypermethylation in AML1/ ETO-positive and -negative myeloid leukemia cell lines. Leuk Res, 2007; 31:497–506.
  22. Bezek S, Ujhazy E, Mach M, Navarova J, Dubovicky M. Developmental origin of chronic diseases: toxicological implication. Interdiscip Toxicol, 2008;1:29–31.
  23. Bhaumik D, Scott GK, Schokrpur S, Patil CK, Campisi J, Benz CC. Expression of microRNA-146 suppresses NF-kappa B activity with reduction of metastatic potential in breast cancer cells. Oncogene, 2008; 27:5643–5647.
  24. Bird A. Perceptions of epigenetics. Nature 2007:447: 396–398.
  25. Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002:16:6–21.
  26. Bloomston M, Frankel WL, Petrocca F et al. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA, 2007; 297:1901–08.
  27. Bollati V, Baccarelli A, Hou L et al. Changes in DNA methylation patterns in subjects exposed to low-dose benzene. Cancer Res, 2007; 67:876–80.
  28. Bollati V, Marinelli B, Apostoli P et al. Exposure to metal-rich particulate matter modifies the expression of candidate microRNAs in peripheral blood leukocytes. Environ Health Perspect, 2010; 118:763–68.
  29. Boonchai W, Walsh M, Cummings M, ChenevixTrench G. Expression of p53 in arsenic-related and sporadic basal cell carcinoma. Arch Dermatol, 2000; 136: 195–98.
  30. Broday L, Peng W, Kuo MH, Salnikov K, Zoroddu M, Costa M. Nickel compounds are novel inhibitors of histone H4 acetylation. Cancer Res, 2000; 60:238–41.
  31. Bromer JG, Zhou Y, Taylor MB, Doherty L, Taylor HS. Bisphenol-A exposure in utero leads to epigenetic alterations in the developmental programming of uterine estrogen response. FASEB J, 2010; 24:2273–80.
  32. Brothman AR, Swanson G, Maxwell TM et al. Global hypomethylation is common in prostate cancer cells: a quantitative predictor for clinical outcome? Cancer Genet Cytogenet, 2005; 156:31–36.
  33. Buckingham L, Penfield Faber L, Kim A et al. PTEN, RASSF1 and DAPK site-specific hypermethylation and outcome in surgically treated stage I and II nonsmall cell lung cancer patients. Int J Cancer, 2010; 126:1630–39.
  34. Budhu A, Jia HL, Forgues M et al. Identification of metastasis-related microRNAs in hepatocellular carcinoma. Hepatology, 2008; 47:897–907.
  35. Calin GA, Dumitru CD, Shimizu M et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA 2002; 99: 15524–29.
  36. Calin GA, Ferracin M, Cimmino A et al. A microRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. New Engl J Med 2005; 353: 1793–801.
  37. Calin GA, Liu CG, Sevignani C et al. MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proc Natl Acad Sci USA, 2004; 101:11755–60.
  38. Cantone L, Nordio F, Hou L et al. Effects of Inhalable Metal-rich Air Particles on Histone H3K4 Dimethylation and H3K9 Acetylation in a Cross-sectional Study of Steel Workers. Environ Health Perspect, 2011; 119:964–9.
  39. Cao Y, Yu SL, Wang Y, Guo GY, Ding Q, An RH. MicroRNA-dependent regulation of PTEN after arsenic trioxide treatment in bladder cancer cell line T24. Tumour Biol, 2011; 32:179–88.
  40. Chai CY, Huang YC, Hung WC, Kang WY, Chen WT. Arsenic salts induced autophagic cell death and hypermethylation of DAPK promoter in SV-40 immortalized human uroepithelial cells. Toxicol Lett, 2007; 173:48–56.
  41. Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res, 2005; 65:6029–33.
  42. Chanda S, Dasgupta UB, Guhamazumder D et al. DNA hypermethylation of promoter of gene p53 and p16 in arsenic-exposed people with and without malignancy. Toxicol Sci, 2006; 89:431–37.
  43. Cheetham S, Tang MJ, Mesak F, Kennecke H, Owen D, Tai IT. SPARC promoter hypermethylation in colorectal cancers can be reversed by 5-Aza-20 deoxycytidine to increase SPARC expression and improve therapy response. British J Cancer 2008; 9 8:1810–19.
  44. Chen H, Ke Q, Kluz T, Yan Y, Costa M. Nickel ions increase histone H3 lysine 9 dimethylation and induce transgene silencing. Mol Cell Biol, 2006; 26:3728–37.
  45. Chen H, Li S, Liu J, Diwan BA, Barrett JC, Waalkes MP. Chronic inorganic arsenic exposure induces hepatic global and individual gene hypomethylation: implications for arsenic hepatocarcinogenesis.Carcinogenesis 2004; 25: 1779–86.
  46. Chen H, Liu J, Zhao CQ, Diwan BA, Merrick BA, Waalkes MP. Association of c-myc overexpression and hyperproliferation with arsenite-induced malignant transformation. Toxicol Appl Pharmacol 2001; 175: 260–68.
  47. Chen MN, Mao Q, Liu YH, Mao BY. Methylation and expression analysis of p16 (INK4a) and RB genes in meningiomas. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 2004; 21:277–79.
  48. Chen MW, Hua KT, Kao HJ et al. H3K9 Histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM. Cancer Res, 2010; 70:7830–40.
  49. Chen W, Wu Y, Zhu J, Liu J, Tan S, Xia C. Methylation of p16 and p15 genes in multiple myeloma. Clin Med Sci J.2002; 17:101–05.
  50. Chen WT, Hung WC, Kang WY, Huang YC, Chai CY. Urothelial carcinomas arising in arsenic-contaminated areas are associated with hypermethylation of the gene promoter of the death-associated protein kinase. Histopathology, 2007; 51:785–92.
  51. Cheng YH, Ji RR, Yue JM et al. MicroRNAs are aberrantly expressed in hypertrophic heart - Do they play a role in cardiac hypertrophy? Am J Pathol 2007; 170: 1831–40.
  52. Chim CS, Fung TK, Liang R. Disruption of INK4/ CDK/Rb cell cycle pathway by gene hypermethylation in multiple myeloma and MGUS. Leukemia 2003; 17: 2533–35.
  53. Chopra M, Amor DJ, Sutton L, Algar E, Mowat D. Russell-Silver syndrome due to paternal H19/IGF2 hypomethylation in a patient conceived using intracytoplasmic sperm injection. Reprod Biomed Online 2010; 20: 843–47.
  54. Cimmino A, Calin GA, Fabbri M et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA2005; 102:13944–49.
  55. Coffin JC, Ge R, Yang S, Kramer PM, Tao L, Pereira MA. Effect of trihalomethanes on cell proliferation and DNA methylation in female B6C3F1 mouse liver. Toxicol Sci, 2000; 58:243–52.
  56. Connolly E, Melegari M, Landgraf P et al. Elevated expression of the miR-17-92 polycistron and miR-21 in hepadnavirus-associated hepatocellular carcinoma contributes to the malignant phenotype. Am J Pathol 2008; 173:856-64.
  57. Coppin JF, Qu W, Waalkes MP. Interplay between cellular methyl metabolism and adaptive efflux during oncogenic transformation from chronic arsenic exposure in human cells. J Biol Chem, 2008; 283:19342–50.
  58. Cui H, Onyango P, Brandenburg S, Wu Y, Hsieh CL, Feinberg AP. Loss of imprinting in colorectal cancer linked to hypomethylation of H19 and IGF2. Cancer Res, 2002;62:6442–46.
  59. Cui X, Wakai T, Shirai Y, Hatakeyama K, Hirano S. Chronic oral exposure to inorganic arsenate interferes with methylation status of p16INK4a and RASSF1A and induces lung cancer in A/J mice. Toxicol Sci, 2006; 91:372–81.
  60. Dammann RH, Kirsch S, Schagdarsurengin U et al. Frequent aberrant methylation of the imprinted IGF2/ H19 locus and LINE1 hypomethylation in ovarian carcinoma. Int J Oncol, 2010; 36:171–79.
  61. Del Senno L, Maestri I, Piva R et al. Differential hypomethylation of the c-myc protooncogene in bladder cancers at different stages and grades. J Urol, 1989; 142:146-49
  62. Deng G, Nguyen A, Tanaka H et al. Regional hypermethylation and global hypomethylation are associated with altered chromatin conformation and histone acetylation in colorectal cancer. Int J Cancer 2006; 118: 2999–3005.
  63. Dixon-McIver A, East P, Mein CA et al. Distinctive patterns of microrna expression associated with karyotype in acute myeloid leukaemia. PLos One 2008; 3:e2141.
  64. Dolinoy DC, Huang D, Jirtle RL. Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development. Proc Natl Acad Sci USA, 2007; 104:13056–61.
  65. El-Shakankiry NH, Mossallam GI. p15 (INK4B) and E-cadherin CpG island methylation is frequent in Egyptian acute myeloid leukemia. J Egypt Natl Canc Inst2006; 18:227–32.
  66. Elyakim E, Sitbon E, Faerman A et al. hsa-miR-191 is a candidate oncogene target for hepatocellular carcinoma therapy. Cancer Res, 2010; 70:8077–87.
  67. Esteller M. Epigenetics in cancer. N Engl J Med, 2008; 358:1148–59.
  68. Fabbri M. et al. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. 2007:104:15805–15810.
  69. Fang JY, Zhu SS, Xiao SD et al. Studies on the hypomethylation of c-myc, c-Ha-ras oncogenes and histopathological changes in human gastric carcinoma. J Gastroenterol Hepatol, 1996; 11:1079–82.
  70. Felli N, Fontana L, Pelosi E et al. MicroRNAs 221 and 222 inhibit normal erythropoiesis and erythroleukemic cell growth via kit receptor down-modulation. Proc Natl Acad Sci USA, 2005; 102:18081–86.
  71. Fradin D & Bougneres P. T2DM: Why Epigenetics? Journal of Nutrition and Metabolism2011:647514. doi:10.1155/2011/647514.
  72. Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A, Lund AH. Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem 2008; 283: 1026–33.
  73. Friedman JMet al. The putative tumor suppressor microRNA- 101 modulates the cancer epigenome by repressing the polycomb group protein EZH2. Cancer Res. 2009:69:2623–2629.
  74. Fu HY, Shen JZ. Hypermethylation of CpG Island of p16 gene and arsenic trioxide induced p16 gene demethylation in multiple myeloma. Zhonghua Nei Ke Za Zhi 2005; 44: 411–14.
  75. Furonaka O, Takeshima Y, Awaya H, Ishida H, Kohno N, Inai K. Aberrant methylation of p14(ARF), p15(INK4b) and p16(INK4a) genes and location of the primary site in pulmonary squamous cell carcinoma. Pathol Int 2004; 54:549–55.
  76. Futscher BW et al. Role for DNA methylation in the control of cell type specific maspin expression. Nat. Genet. 2002:31:175–179.
  77. Gaikwad AB, Sayyed SG, Lichtnekert J, Tikoo K, Anders HJ. Renal failure increases cardiac histone h3 acetylation, dimethylation, and phosphorylation and the induction of cardiomyopathy-related genes in type 2 diabetes. Am J Pathol, 2010; 176:1079–83.
  78. Gallardo F, Esteller M, Pujol RM, Costa C, Estrach T, Servitje O. Methylation status of the p15, p16 and MGMT promoter genes in primary cutaneous T-cell lymphomas. Haematologica, 2004; 89:1401–403.
  79. Gal-Yam, EN et al. Constitutive nucleosome depletion and ordered factor assembly at the GRP78 promoter revealed by single molecule footprinting. Plos Genet. 2006:2:e160.
  80. Gao A, Zuo X, Liu Q, Lu X, Guo W, Tian L. Methylation of PARP-1 promoter involved in the regulation of benzene-induced decrease of PARP-1 mRNA expression. Toxicol Lett, 2010; 195:114–18.
  81. Gao WM, Chadha MS, Kline AE et al. Immunohistochemical analysis of histone H3 acetylation and methylationevidence for altered epigeneticsignaling following traumatic brain injury in immature rats. Brain Res, 2006; 1070:31–4.
  82. Garofalo M, Di Leva G, Romano G et al. miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell, 2009; 16:498–509.
  83. Geneletti S, Gallo V, Porta M, Khoury MJ, Vineis P. Assessing causal relationships in genomics: From Bradford-Hill criteria to complex gene-environment interactions and directed acyclic graphs. Emerg Themes Epidemiol, 2011; 8:5–22.
  84. Golebiowski F, Kasprzak KS. Inhibition of core histones acetylation by carcinogenic nickel (II). Mol Cell Biochem, 2005; 279:133–39.
  85. Gonzalez-Ramirez I, Ramirez-Amador V, IrigoyenCamacho ME et al. hMLH1 promoter methylation is an early event in oral cancer. Oral Oncol, 2011; 47:22–26.
  86. Govindarajan B, Klafter R, Miller MS et al. Reactive oxygen-induced carcinogenesis causes hypermethylation of p16 (Ink4a) and activation of MAP kinase. Mol Med, 2002; 8:1–8.
  87. Gramantieri L, Ferracin M, Fornari F et al. Cyclin g1 is a target of miR-122a, MicroRNA frequently down-regulated in human hepatocellular carcinoma. Cancer Res, 2007; 67: 6092–99.
  88. Grewal SI, Moazed D. Heterochromatin and epigenetic control of gene expression. Science 2003:301:798–802.
  89. Gronbaek K, Ralfkiaer U, Dahl C et al. Frequent hypermethylation of DBC1 in malignant lymphoproliferative neoplasms. Mod Pathol, 2008;21:632–38.
  90. Gucev ZS, Tasic V, Jancevska A, Kirovski I. A case of Silver-Russell syndrome (SRS): multiple pituitary hormone deficiency, lack of H19 hypomethylation and favourable growth hormone (GH) treatment response. J Genet, 2009;88:239–43.
  91. Guerrero-Bosagna C, Settles M, Lucker B, Skinner MK. Epigenetic transgenerational actions of vinclozolin on promoter regions of the sperm epigenome. PLoS One 2010; 5:e13100.
  92. Hafner N, Diebolder H, Jansen L, Hoppe I, Durst M, Runnebaum IB. Hypermethylated DAPK in serum DNA of women with uterine leiomyoma is a biomarker not restricted to cancer. Gynecol Oncol, 2011; 121:224–9.
  93. Hanafusa T, Shinji T, Shiraha H et al. Functional promoter upstream p53 regulatory sequence of IGFBP3 that is silenced by tumor specific methylation. BMC Cancer, 2005; 5:9–19.
  94. Harikrishnan KN et al. Brahma links the SWI/SNF chromatin remodeling complex with MeCP2-dependent transcriptional silencing. Nat. Genet. 2005:37:254–264.
  95. Hattori N et al. Epigenetic control of mouse Oct-4 gene expression in embryonic stem cells and trophoblast stem cells. J. Biol. Chem.2004:279:17063–17069.
  96. Hayashita Y, Osada H, Tatematsu Y et al. A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res, 2005;65:9628–32.
  97. He L et al. MicroRNAs: small RNAs with a big role in gene regulation. Nat. Rev. Genet.2004;5:522–531.
  98. Hebert C, Norris K, Scheper MA, Nikitakis N, Sauk JJ. High mobility group A2 is a target for miRNA-98 in head and neck squamous cell carcinoma. Mol Cancer 2007;6: 5–15.
  99. Hebert SS, Horre K, Nicolai L et al. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer’s disease correlates with increased BACE1/beta-secretase expression. Proc Natl Acad Sci USA2008; 105:6415–20.
  100. Heightman TD. Therapeutic prospects for epigenetic modulation. Expert Opin Ther Targets2011;15:729–40.
  101. Heng JI, Nguyen L, Castro DS et al. Neurogenin 2 controls cortical neuron migration through regulation of Rnd2. Nature, 2008; 455:114–18.
  102. Hill VK, Hesson LB, Dansranjavin T et al. Identification of 5 novel genes methylated in breast and other epithelial cancers. Mol Cancer, 2010; 9:51–63.
  103. Hou L, Zhang X, Wang D & Baccarelli, A. Environmental chemical exposures and human epigenetics. International Journal of Epidemiology, 2012; 41:79–105. http://doi.org/10.1093/ije/dyr154
  104. Hsu PY, Deatherage DE, Rodriguez BA et al. Xenoestrogeninduced epigenetic repression of microRNA- 9-3 in breast epithelial cells. Cancer Res, 2009; 69:5936–45.
  105. Hu SL, Kong XY, Cheng ZD et al. Promoter methylation of p16, Runx3, DAPK and CHFR genes is frequent in gastric carcinoma. Tumori, 2010; 96:726–33.
  106. Huang D, Zhang Y, Qi Y, Chen C, Ji W. Global DNA hypomethylation, rather than reactive oxygen species (ROS), a potential facilitator of cadmium-stimulated K562 cell proliferation. Toxicol Lett, 2008; 179:43–47.
  107. Iorio MV, Ferracin M, Liu CG et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res, 2005; 65:7065–70.
  108. Ito Y, Koessler T, Ibrahim AE et al. Somatically acquired hypomethylation of IGF2 in breast and colorectal cancer. Hum Mol Genet, 2008; 17:2633–43.
  109. Jazdzewski K, Murray EL, Franssila K, Jarzab B, Schoenberg DR, de la Chapelle A. Common SNP in pre-miR-146a decreases mature miR expression and predisposes to papillary thyroid carcinoma. Proc Natl Acad Sci USA, 2008; 105:7269–74.
  110. Jensen TJ, Novak P, Eblin KE, Gandolfi AJ, Futscher BW. Epigenetic remodeling during arsenical-induced malignant transformation. Carcinogenesis, 2008; 29:1500–508.
  111. Jenuwein, T. et al. Translating the histone code. Science, 2001; 293:1074–1080.
  112. Ji Z, Zhang L, Peng V, Ren X, McHale CM, Smith MT. A comparison of the cytogenetic alterations and global DNA hypomethylation induced by the benzene metabolite, hydroquinone, with those induced by melphalan and etoposide. Leukemia, 2010; 24:986–91.
  113. Jiang, C. et al. Nucleosome positioning and gene regulation: advances through genomics. Nat. Rev. Genet. 2009; 10: 161–172.
  114. Jo WJ, Ren XF, Chu FX et al. Acetylated H4K16 by MYST1 protects UROtsa cells from arsenic toxicity and is decreased following chronic arsenic exposure. Toxicol Appl Pharmacol2009; 241:294–302.
  115. Johnson SM, Grosshans H, Shingara J et al. RAS is regulated by the let-7 MicroRNA family. Cell 2005; 120: 635–47.
  116. Jones, P.L. et al. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat. Genet. 1998; 19:187–191.
  117. Kanao K, Mikami S, Mizuno R, Shinojima T, Murai M, Oya M. Decreased acetylation of histone H3 in renal cell carcinoma: a potential target of histone deacetylase inhibitors. J Urol 2008; 180:1131–36.
  118. Karaczyn A, Ivanov S, Reynolds M, Zhitkovich A, Kasprzak KS, Salnikow K. Ascorbate depletion mediates up-regulation of hypoxia-associated proteins by cell density and nickel. J Cell Biochem, 2006; 97:1025–35.
  119. Karaczyn AA, Golebiowski F, Kasprzak KS. Truncation, deamidation, and oxidation of histone H2B in cells cultured with nickel (II). Chem Res Toxicol 2005; 18:1934–42.
  120. Ke Q, Davidson T, Chen H, Kluz T, Costa M. Alterations of histone modifications and transgene silencing by nickel chloride. Carcinogenesis, 2006; 27:1481–88.
  121. Ke Q, Li Q, Ellen TP, Sun H, Costa M. Nickel compounds induce phosphorylation of histone H3 at serine 10 by activating JNK-MAPK pathway. Carcinogenesis 2008; 29: 1276–81.
  122. Ke QD, Davidson T, Chen HB, Kluz T, Costa M. Alterations of histone modifications and transgene silencing by nickel chloride. Carcinogenesis, 2006; 27:1481–88.
  123. Kim KY, Kim DS, Lee SK et al. Association of low-dose exposure to persistent organic pollutants with global DNA hypomethylation in healthy Koreans. Environ Health Perspect, 2010; 118:370–74.
  124. Kim M, Yim SH, Cho NS et al. Homozygous deletion of CDKN2A (p16, p14) and CDKN2B (p15) genes is a poor prognostic factor in adult but not in childhood B-lineage acute lymphoblastic leukemia: a comparative deletion and hypermethylation study. Cancer Genet Cytogenet, 2009; 195:59–65.
  125. Klein CB, Costa M. DNA methylation, heterochromatin and epigenetic carcinogens. Mutat Res-Rev Mutat Res 1997; 386:163–80.
  126. Klein CB, Su L, Bowser D, Leszczynska J. Chromate-induced epimutations in mammalian cells. Environ Health Perspect 2002; 110 (Suppl 5):739–43.
  127. Kondo K, Takahashi Y, Hirose Y et al. The reduced expression and aberrant methylation of p16 (INK4a) in chromate workers with lung cancer. Lung Cancer 2006; 53:295–302.
  128. Koutros S, Alavanja MC, Lubin JH et al. An update of cancer incidence in the Agricultural Health Study. J Occup Environ Med, 2010;52:1098–105.
  129. Kouzarides T. Chromatin modifications and their function. Cell, 2007; 128:693–705.
  130. Kozaki KI, Imoto I, Mogi S, Omura K, Inazawa J. Exploration of tumor-suppressive microRNAs silenced by DNA hypermethylation in oral cancer. Cancer Res, 2008;68:2094–105.
  131. Kumar R, Hunt CR, Gupta A et al. Purkinje cell-specific males absent on the first (mMof) gene deletion results in an ataxia-telangiectasia-like neurological phenotypeand backward walking in mice. Proc Natl Acad Sci USA, 2011;108:3636–41.
  132. Laytragoon-Lewin N, Chen F, Castro J et al. DNA content and methylation of p16, DAPK and RASSF1A gene in tumour and distant, normal mucosal tissue of head and neck squamous cell carcinoma patients. Anticancer Res, 2010;30:4643–48.
  133. le Sage C, Nagel R, Egan DA et al. Regulation of the p27(Kip1) tumor suppressor by miR-221 and miR-222 promotes cancer cell proliferation. EMBO J 2007;26: 3699–708.
  134. Lee YS, Dutta A. The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. Genes Dev 2007;21: 1025–30.
  135. Lee YW, Klein CB, Kargacin B et al. Carcinogenic nickel silences gene-expression by chromatin condensation and DNA methylation - a new model for epigenetic carcinogens. Mol Cell Biol, 1995;15:2547–57.
  136. Lefevre GM, Patel SR, Kim D, Tessarollo L, Dressler GR. Altering a histone H3K4 methylation pathway in glomerular podocytes promotes a chronic disease phenotype. PLoS Genet 2010;6:e1001142.
  137. Lelieveld J, Evans JS, Fnais M, Giannadaki D& Pozzer A. The contribution of outdoor air pollution sources to premature mortality on a global scale. Nature 2015;525(7569):367–371. Retrieved from http://dx.doi.org/10.1038/nature15371
  138. Li B, Wang B, Niu LJ, Jiang L, Qiu CC. Hypermethylation of multiple tumor-related genes associated with DMNT3b up-regulation served as a biomarker for early diagnosis of esophageal squamous cell carcinoma. Epigenetics, 2011;6:307–16.
  139. Li H, Lu S, Fong L. Study on the status of methylation of Rb gene promoter in human esophageal cancer and effect of NMBzA on Rb gene promoter in monkey esophageal epithelium. Zhonghua Zhong Liu Za Zhi, 1998;20:412–14.
  140. Li J, Chen P, Sinogeeva N et al. Arsenic trioxide promotes histone H3 phosphoacetylation at the chromatin of CASPASE-10 in acute promyelocytic leukemia cells. J Biol Chem 2002;277:49504–10.
  141. Li J, Gorospe M, Barnes J, Liu Y. Tumor promoter arsenite stimulates histone H3 phosphoacetylation of proto-oncogenes c-fos and c-jun chromatin in human diploid fibroblasts. J Biol Chem 2003;278: 13183–91.
  142. Li Y, Meng G, Huang L, Guo QN. Hypomethylation of the P3 promoter is associated with up-regulation of IGF2 expression in human osteosarcoma. Hum Pathol 2009;40: 1441–47.
  143. Lin HH, Ke HL, Wu WJ, Lee YH, Chang LL. Hypermethylation of E-cadherin, p16, p14, and RASSF1A genes in pathologically normal urothelium predict bladder recurrence of bladder cancer after transurethral resection. Urologic Oncology: Seminars and Original Investigations 2012;30(2): 177–181.
  144. Lin SL, Chiang A, Chang D, Ying SY. Loss of mir-146a function in hormone-refractory prostate cancer. Rna-a Publication of the RNA Societym 2008;14:417–24.
  145. Lin JC et al. Role of nucleosomal occupancy in the epigenetic silencing of the MLH1 CpG Island. Cancer Cell, 2007;12:432–444.
  146. Lindberg D, Akerstrom G, Westin G. Evaluation of CDKN2C/p18, CDKN1B/p27 and CDKN2B/p15 mRNA expression, and CpG methylation status in sporadic and MEN1-associated pancreatic endocrine tumours. Clin Endocrinol, 2008;68:271–77.
  147. Ling ZQ, Tanaka A, Li P et al. Microsatellite instability with promoter methylation and silencing of hMLH1 can regionally occur during progression of gastric carcinoma. Cancer Lett, 2010;297:244–51.
  148. Liu J, Ballaney M, Al-alem U et al. Combined inhaled diesel exhaust particles and allergen exposure alter methylation of T helper genes and IgE production in vivo. Toxicol Sci, 2008;102:76–81.
  149. Luger Ket al. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature, 1997;389:251–260.
  150. Lukiw WJ, Pogue AI. Induction of specific micro RNA (miRNA) species by ROS-generating metal sulfates in primary human brain cells. J Inorg Biochem 2007;101: 1265–69.
  151. Lukiw WJ, Zhao YH, Cui JG. An NF-kappa B-sensitive micro RNA-146a-mediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J Biol Chem, 2008;283:31315–22.
  152. Lukiw WJ. Micro-RNA speciation in fetal, adult and Alzheimer’s disease hippocampus. Neuroreport 2007;18: 297–300.
  153. Luo J, Li YN, Wang F, Zhang WM, Geng X. S-adenosylmethionine inhibits the growth of cancer cells by reversing the hypomethylation status of c-myc and H-ras in human gastric cancer and colon cancer. Int J Biol Sci, 2010;6:784–95.
  154. Majumdar S, Chanda S, Ganguli B, Mazumder DN, Lahiri S, Dasgupta UB. Arsenic exposure induces genomic hypermethylation. Environ Toxicol, 2010;25:315–18.
  155. Malhotra P, Kochhar R, Vaiphei K, Wig JD, Mahmood S. Aberrant promoter methylation of p16 in colorectal adenocarcinoma in North Indian patients. World J Gastrointest Oncol, 2010;2:295–303.
  156. Malzkorn B, Wolter M, Grzendowski M, Stuhler K, Reifenberger G. Identification and functional characterization of microRNAs involved in the malignant progression of astrocytic gliomas. Brain Pathol, 2010;20:539–50.
  157. Marsit CJ, Karagas MR, Danaee H et al. Carcinogen exposure and gene promoter hypermethylation in bladder cancer. Carcinogenesis, 2006;27:112–16.
  158. Mass MJ, Wang L. Arsenic alters cytosine methylation patterns of the promoter of the tumor suppressor gene p53 in human lung cells: a model for a mechanism of carcinogenesis. Mutat Res, 1997;386:263–77.
  159. Matsuno N, Hoshino K, Nanri T et al. p15 mRNA expression detected by real-time quantitative reverse transcriptase-polymerase chain reaction correlates with the methylation density of the gene in adult acute leukemia. Leuk Res, 2005;29:557–64.
  160. McClure EA, North CM, Kaminski NE, Goodman JI. Changes in DNA methylation and gene expression during 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)induced suppression of the lipopolysaccharide (LPS)stimulated IgM response in splenocytes. Toxicol Sci, 2011;120:339–48.
  161. Meng FY, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007;133: 647–58.
  162. Mi SL, Lu J, Sun M et al. MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia. Blood, 2007;110:75a.
  163. Mill J, Tang T, Kaminsky Z et al. Epigenomic profiling reveals DNA-methylation changes associated with major psychosis. Am J Hum Genet, 2008;82:696–711.
  164. Minard ME, Jain AK, Barton MC. Analysis of epigenetic alterations to chromatin during development. Genesis, 2009;47:559–72.
  165. Minardi D, Lucarini G, Filosa A et al. Prognostic role of global DNA-methylation and histone acetylation in pT1a clear cell renal carcinoma in partial nephrectomy specimens. J Cell Mol Med, 2009;13:2115–21.
  166. Morgan DA, Rahmouni K. Differential effects of insulin on sympathetic nerve activity in agouti obese mice. J Hypertens, 2010;28:1913–19.
  167. Nan X et al. Transcriptional repression by the methyl-CpGbinding protein MeCP2 involves a histone deacetylase complex. Nature, 1998;393:386–389.
  168. Norrie MWA, Hawkins NJ, Todd AV, Meagher AP, O’Connor TW, Ward RL. The role of hMLH1 methylation in the development of synchronous sporadic colorectal carcinomas. Dis Colon Rectum, 2002;45:674–80.
  169. Okoji RS, Yu RC, Maronpot RR, Froines JR. Sodium arsenite administration via drinking water increases genome-wide and Ha-ras DNA hypomethylation in methyl-deficient C57BL/6J mice. Carcinogenesis, 2002;23:777–85.
  170. Owen HC, Giedl J, Wild PJ et al. Low frequency of epigenetic events in urothelial tumors in young patients. J Urol, 2010;184:459–63.
  171. Paluszczak J, Misiak P, Wierzbicka M, Wozniak A, BaerDubowska W. Frequent hypermethylation of DAPK, RARbeta, MGMT, RASSF1A and FHIT in laryngeal squamous cell carcinomas and adjacent normal mucosa. Oral Oncol, 2011;47:104–7.
  172. Paul TA, Bies J, Small D, Wolff L. Signatures of polycomb repression and reduced H3K4 trimethylation are associated with p15INK4b DNA methylation in AML. Blood, 2010;115:3098–108.
  173. Peng Z, Shan C, Wang H. Value of promoter methylation of RASSF1A, p16, and DAPK genes in induced sputum in diagnosing lung cancers. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2010;35:247–53.
  174. Pereira MA, Kramer PM, Conran PB, Tao L. Effect of chloroform on dichloroacetic acid and trichloroacetic acid-induced hypomethylation and expression of the c-myc gene and on their promotion of liver and kidney tumors in mice. Carcinogenesis, 2001;22:1511–19.
  175. Pereira MA, Tao L, Liu Y, Li L, Steele VE, Lubet RA. Modulation by budesonide of DNA methylation and mRNA expression in mouse lung tumors. Int J Cancer, 2007;120:1150–53.
  176. Pilsner JR, Hu H, Ettinger A et al. Influence of prenatal lead exposure on genomic methylation of cord blood DNA. Environ Health Perspect, 2009;117:1466–71.
  177. Pilsner JR, Lazarus AL, Nam DH et al. Mercury-associated DNA hypomethylation in polar bear brains via the Luminometric Methylation Assay: a sensitive method to study epigenetics in wildlife. Mol Ecol, 2010;19:307–14.
  178. Pilsner JR, Liu X, Ahsan H et al. Folate deficiency, hyperhomocysteinemia, low urinary creatinine, and hypomethylation of leukocyte DNA are risk factors for arsenic-induced skin lesions. Environ Health Perspect, 2009;117:254–60.
  179. Pilsner JR, Liu X, Ahsan H et al. Genomic methylation of peripheral blood leukocyte DNA: influences of arsenic and folate in Bangladeshi adults. Am J Clin Nutr 2007; 86: 1179–86.
  180. Poetsch M, Hemmerich M, Kakies C et al. Alterations in the tumor suppressor gene p16 (INK4A) are associated with aggressive behavior of penile carcinomas. Virchows Arch, 2011;458:221–29.
  181. Pogue AI, Li YY, Cui JG et al. Characterization of an NF-kappa B-regulated, miRNA-146a-mediated downregulation of complement factor H (CFH) in metalsulfate-stressed human brain cells. J Inorg Biochem, 2009;103:1591–95.
  182. Porcedda P, Turinetto V, Brusco A et al. A rapid flow cytometry test based on histone H2AX phosphorylation for the sensitive and specific diagnosis of ataxia telangiectasia. Cytometry A2008;73:508–16.
  183. Prendergast GC et al. Methylation-sensitive sequence-specific DNA binding by the c-Myc basic region. Science, 1991;251:186–189.
  184. Pru¨ss-U¨stu¨n Annette CC. Preventing disease through healthy environments. Towards an estimate of the environmental burden of disease World Health Organization (WHO), 2006.
  185. Qian J, Yao DM, Lin J et al. Alteration of methylation status of death-associated protein kinase (dapk) gene promoter in patients with acute myeloid leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2010;18: 1390–94.
  186. Rabiau N, Thiam MO, Satih S et al. Methylation analysis of BRCA1, RASSF1, GSTP1 and EPHB2 promoters in prostate biopsies according to different degrees of malignancy. In Vivo, 2009;23:387–91.
  187. Radpour R, Barekati Z, Haghighi MM et al. Correlation of telomere length shortening with promoter methylation profile of p16/Rb and p53/p21 pathways in breast cancer. Mod Pathol, 2010;23:763–72.
  188. Ramirez T, Brocher J, Stopper H, Hock R. Sodium arsenite modulates histone acetylation, histone deacetylase activity and HMGN protein dynamics in human cells. Chromosoma, 2008;117:147–57.
  189. Reichard JF, Schnekenburger M, Puga A. Long term low-dose arsenic exposure induces loss of DNA methylation. Biochem Biophys Res Commun2007;352:188–92.
  190. Reik W, Dean W, Walter J. Epigenetic reprogramming in mammalian development. Science2001;293:1089–93.
  191. Relton CL, Davey Smith G. Epigenetic epidemiology of common complex disease: prospects for prediction, prevention, and treatment. PLoS Med 2010;7:e1000356.
  192. Roccaro AM, Jia X, Ngo HT et al. MicroRNA expression in the biology, prognosis, and therapy of waldenstrom macroglobulinemia. Clin Lymphoma Myeloma 2009;9: S126.
  193. Roccaro AM, Sacco A, Thompson B et al. MicroRNAs 15a and 16 regulate tumor proliferation in multiple myeloma. Blood, 2009;113:6669–80.
  194. Roehle A, Hoefig KP, Repsilber D et al. MicroRNA signatures characterize diffuse large B-cell lymphomas and follicular lymphomas. British J Haematol 2008;142: 732–44.
  195. Roman-Gomez J, Jimenez-Velasco A, Agirre X et al. Promoter hypermethylation and global hypomethylation are independent epigenetic events in lymphoid leukemogenesis with opposing effects on clinical outcome. Leukemia, 2006;20:1445–48.
  196. Rusiecki JA, Baccarelli A, Bollati V, Tarantini L, Moore LE, Bonefeld-Jorgensen EC. Global DNA hypomethylation is associated with high serum-persistent organic pollutants in Greenlandic Inuit. Environ Health Perspect, 2008;116: 1547–52.
  197. Saba R, Goodman CD, Huzarewich RLCH, Robertson C, Booth SA. A miRNA Signature of Prion Induced Neurodegeneration. PLos One 2008;3:e3652.
  198. Saito Y. et al. Epigenetic activation of tumor suppressor micro- RNAs in human cancer cells. Cell Cycle, 2006;5:2220–2222.
  199. Sampson VB, Rong NH, Han J et al. MicroRNA let-7a down-regulates MYC and reverts MYC-induced growth in Burkitt lymphoma cells. Cancer Res 2007;67: 9762–70.
  200. Saumet A, Vetter G, Bouttier M et al. Transcriptional repression of microRNA genes by PML-RARA increases expression of key cancer proteins in acute promyelocytic leukemia. Blood, 2009;113:412–21.
  201. Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M. MicroRNAs play an essential role in the development of cardiac hypertrophy. Circulation Res, 2007;100:416–24.
  202. Sayyed SG, Gaikwad AB, Lichtnekert J et al. Progressive glomerulosclerosis in type 2 diabetes is associated with renal histone H3K9 and H3K23 acetylation, H3K4 dimethylation and phosphorylation at serine 10. Nephrol Dial Transplant, 2010; 25:1811–17.
  203. Schafer E, Irizarry R, Negi S et al. Promoter hypermethylation in MLL-r infant acute lymphoblastic leukemia: biology and therapeutic targeting. Blood 2010; 115: 4798–809.
  204. Schetter AJ, Leung SY, Sohn JJ et al. MicroRNA expression profiles associated with prognosis and therapeuticoutcome in colon adenocarcinoma. JAMA 2008; 299: 425–36.
  205. Schones, D.E. et al. (2008) Dynamic regulation of nucleosome positioning in the human genome. Cell, 132, 887–898.
  206. Sciandrello G, Caradonna F, Mauro M, Barbata G. Arsenic-induced DNA hypomethylation affects chromosomal instability in mammalian cells. Carcinogenesis 2004; 25:413–17.
  207. Seligson DB, Horvath S, McBrian MA et al. Global Levels of Histone Modifications Predict Prognosis in Different Cancers. Am J Pathol, 2009; 174:1619–28.
  208. Serizawa RR, Ralfkiaer U, Steven K et al. Integrated genetic and epigenetic analysis of bladder cancer reveals an additive diagnostic value of FGFR3 mutations and hypermethylation events. Int J Cancer, 2011; 129:78–87.
  209. Sharma S, Kelly TK & Jones PA. Epigenetics in cancer. Carcinogenesis, 2010; 31(1):27–36. http://doi.org/10.1093/carcin/bgp220
  210. Shaw R, Risk JM, Liloglou T. RE: P16 INK41 promoter hypermethylation is associated with invasiveness and prognosis of oral squamous cell carcinoma in an age dependent manner. Su et al. Oral Oncol, 2010; 46:734–39.
  211. Shen J, Ambrosone CB, DiCioccio RA, Odunsi K, Lele SB, Zhao H. A functional polymorphism in the miR-146a gene and age of familial breast/ovarian cancer diagnosis. Carcinogenesis, 2008; 29:1963–66.
  212. Shen L, Qui D, Fang J. Correlation between hypomethylation of c-myc and c-N-ras oncogenes and pathological changes in human hepatocellular carcinoma. Zhonghua Zhong Liu Za Zhi, 1997; 19:173–76.
  213. Shimabukuro M, Sasaki T, Imamura A et al. Global hypomethylation of peripheral leukocyte DNA in male patients with schizophrenia: a potential link between epigenetics and schizophrenia. J Psychiatr Res 2007;41: 1042–46.
  214. Shimamoto T, Ohyashiki JH, Ohyashiki K. Methylation of p15(INK4b) and E-cadherin genes is independently correlated with poor prognosis in acute myeloid leukemia. Leuk Res, 2005;29:653–59.
  215. Shivaswamy S et al. Dynamic remodeling of individual nucleosomes across a eukaryotic genome in response to transcriptional perturbation. PLoS Biol. 2008;6:e65.
  216. Smith IM, Mydlarz WK, Mithani SK, Califano JA. DNA global hypomethylation in squamous cell head and neck cancer associated with smoking, alcohol consumption and stage. Int J Cancer, 2007;121:1724–28.
  217. Smith, CL et al. ATP-dependent chromatin remodeling. Curr. Top. Dev. Biol. 2005;65: 115–148.
  218. Song C, Kanthasamy A, Anantharam V, Sun F, Kanthasamy AG. Environmental neurotoxic pesticide increases histone acetylation to promote apoptosis in dopaminergic neuronal cells: relevance to epigenetic mechanisms of neurodegeneration. Mol Pharmacol 2010; 77:621–32.
  219. Stirzaker C, Millar DS, Paul CL et al. Extensive DNA methylation spanning the Rb promoter in retinoblastoma tumors. Cancer Res, 1997; 57:2229–37.
  220. Sugita H, Iida S, Inokuchi M et al. Methylation of BNIP3 and DAPK indicates lower response to chemotherapy and poor prognosis in gastric cancer. Oncol Rep 2011; 25: 513–18.
  221. Suzuki MM et al. DNA methylation landscapes: provocative insights from epigenomics. Nat. Rev. Genet. 2008; 9:465–476.
  222. Taganov KD, Boldin MP, Chang KJ, Baltimore D. NF-kappa B-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA 2006; 103:12481–86.
  223. Takahashi M, Barrett JC, Tsutsui T. Transformation by inorganic arsenic compounds of normal Syrian hamster embryo cells into a neoplastic state in which they become anchorage-independent and cause tumors in newborn hamsters. Int J Cancer, 2002; 99:629–34.
  224. Takahashi Y, Kondo K, Hirose T et al. Microsatellite instability and protein expression of the DNA mismatch repair gene, hMLH1, of lung cancer in chromate-exposed workers. Mol Carcinogenesis, 2005;42:150–58.
  225. Takai Det al. Comprehensive analysis of CpG islands in human chromosomes 21 and 22. Proc. Natl Acad. Sci. USA 2002;99:3740–3745.
  226. Takakura S, Mitsutake N, Nakashima M et al. Oncogenic role of miR-17-92 cluster in anaplastic thyroid cancer cells. Cancer Sci, 2008;99:1147–54.
  227. Takiguchi M, Achanzar WE, Qu W, Li G, Waalkes MP. Effects of cadmium on DNA-(Cytosine-5) methyltransferase activity and DNA methylation status during cadmium-induced cellular transformation. Exp Cell Res, 2003;286:355–65.
  228. Tan HX, Wang QA, Chen LZ et al. MicroRNA-9 reduces cell invasion and E-cadherin secretion in SK-Hep-1 cell. Med Oncol2010;27:654–60.
  229. Tang WY, Ho SM. Epigenetic reprogramming and imprinting in origins of disease. Rev Endocr Metab Disord, 2007;8:173–82.
  230. Tao L, Yang S, Xie M, Kramer PM, Pereira MA. Effect of trichloroethylene and its metabolites, dichloroacetic acid and trichloroacetic acid, on the methylation and expression of c-Jun and c-Myc protooncogenes in mouse liver: prevention by methionine. Toxicol Sci, 2000;54:399–407.
  231. Tao L, Yang S, Xie M, Kramer PM, Pereira MA. Hypomethylation and overexpression of c-jun and c-myc protooncogenes and increased DNA methyltransferase activity in dichloroacetic and trichloroacetic acidpromoted mouse liver tumors. Cancer Lett 2000;158: 185–93.
  232. Tarantini L, Bonzini M, Apostoli P et al. Effects of particulate matter on genomic DNA methylation content and iNOS promoter methylation. Environ Health Perspect, 2009;117:217–22.
  233. Tsujiuchi T, Tsutsumi M, Sasaki Y, Takahama M, Konishi Y. Hypomethylation of CpG sites and c-myc gene overexpression in hepatocellular carcinomas, but not hyperplastic nodules, induced by a choline-deficient L-amino acid-defined diet in rats. Jpn J Cancer Res 1999; 90:909–13.
  234. Uthus EO, Davis C. Dietary arsenic affects dimethylhydrazine-induced aberrant crypt formation and hepatic global DNA methylation and DNA methyltransferase activity in rats. Biol Trace Elem Res 2005;103: 133–45.
  235. Van der Auwera I, Bovie C, Svensson C et al. Quantitative methylation profiling in tumor and matched morphologically normal tissues from breast cancer patients. BMC Cancer, 2010;10:97–104.
  236. Van Rooij E, Sutherland LB, Liu N et al. A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure. Proc Natl Acad Sci USA, 2006;103:18255–60.
  237. Vasavi M, Kiran V, Ravishankar B, Prabhakar B, Ahuja YR, Hasan Q. Microsatellite instability analysis and its correlation with hMLH1 repair gene hypermethylation status in esophageal pathologies including cancers. Cancer Biomark, 2010;7:1–10.
  238. Veerla S, Lindgren D, Kvist A et al. MiRNA expression in urothelial carcinomas: Important roles of miR-10a, miR-222, miR-125b, miR-7 and miR-452 for tumor stage and metastasis, and frequent homozygous losses of miR-31. Int J Cancer, 2009;124:2236–42.
  239. Volinia S, Calin GA, Liu CG et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA, 2006;103:2257–61.
  240. Waalkes MP, Liu J, Chen H et al. Estrogen signaling in livers of male mice with hepatocellular carcinoma induced by exposure to arsenic in utero. J Natl Cancer Inst, 2004;96:466–74.
  241. Waggoner JK, Kullman GJ, Henneberger PK et al. Mortality in the agricultural health study, 1993–2007. Am J Epidemiol, 2011;173:71–83.
  242. Wang D, Wang J, Li Y, He Z, Zhang Y. The influence of anthracosis and p16 ink4a gene aberrant methylation on small-sized pulmonary adenocarcinoma. Exp Mol Pathol, 2011;90:131–36.
  243. Wang J, Zhao YY, Liu H et al. The role of insulin-like growth factor-2 gene differentially methylated regions in TCDD-induced malformation. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 2007;24:162–66.
  244. Wang SC, Oelze B, Schumacher A. Age-specific epigenetic drift in late-onset Alzheimer’s disease. PLoS One 2008; 3:e2698.
  245. Wang Y, Lee ATC, Ma JZI et al. Profiling MicroRNA expression in hepatocellular carcinoma reveals MicroRNA-224 up-regulation and apoptosis inhibitor-5 as a MicroRNA-224-specific target. J Biol Chem 2008; 283:13205–15.
  246. Wang Y et al. An evaluation of new criteria for CpG islands in the human genome as gene markers. Bioinformatics, 2004;20:1170–1177.
  247. Watt F et al. Cytosine methylation prevents binding to DNA of a HeLa cell transcription factor required for optimal expression of the adenovirus major late promoter. Genes Dev. 1988; 2:1136–1143.
  248. Wemmert S, Bettscheider M, Alt S et al. p15 promoter methylation - a novel prognostic marker in glioblastoma patients. Int J Oncol, 2009;34:1743–48.
  249. Weng YI, Hsu PY, Liyanarachchi S et al. Epigenetic influences of low-dose bisphenol A in primary human breast epithelial cells. Toxicol Appl Pharmacol 2010;248: 111–21.
  250. Williams AE, Perry MM, Moschos SA, LarnerSvensson HM, Lindsay MA. Role of miRNA-146a in the regulation of the innate immune response and cancer. Biochem Soc Trans 2008; 36(Pt 6):1211–15.
  251. Wong TS, Man MW, Lam AK, Wei WI, Kwong YL, Yuen AP. The study of p16 and p15 gene methylation in head and neck squamous cell carcinoma and theirquantitative evaluation in plasma by real-time PCR. Eur J Cancer,2003;39:1881–87.
  252. Wright RJ. Epidemiology of stress and asthma: from constricting communities and fragile families to epigenetics. Immunol Allergy Clin North Am, 2011;31:19–39.
  253. Wright RO, Schwartz J, Wright RJ et al. Biomarkers of lead exposure and DNA methylation within retrotransposons. Environ Health Perspect2010; 118:790–95.
  254. WysockaJ et al. A PHD finger of NURF couples histone H3 lysine 4 trimethylation with chromatin remodelling. Nature, 2006;442:86–90.
  255. Xi Y, Formentini A, Chien M et al. Prognostic values of microRNAs in colorectal cancer. Biomark Insights 2006; 2: 113–21.
  256. Xiang Z, Proneth B, Dirain ML, Litherland SA, Haskell-Luevano C. Pharmacological characterization of 30 human melanocortin-4 receptor polymorphisms with the endogenous proopiomelanocortin-derived agonists, synthetic agonists, and the endogenous agoutirelated protein antagonist.Biochemistry, 2010;49:4583–600.
  257. Xie Y, Trouba KJ, Liu J, Waalkes MP, Germolec DR. Biokinetics and subchronic toxic effects of oral arsenite, arsenate, monomethylarsonic acid, and dimethylarsinic acid in v-Ha-ras transgenic (Tg.AC) mice. Environ Health Perspect, 2004; 112:1255–63.
  258. Xu T, Zhu Y, Wei QK et al. A functional polymorphism in the miR-146a gene is associated with the risk for hepatocellular carcinoma. Carcinogenesis, 2008; 29: 2126–31.
  259. Yalcin A, Serin MS, Emekdas G et al. Promoter methylation of P15 (INK4B) gene is possibly associated with parvovirus B19 infection in adult acute leukemias. Int J Lab Hematol, 2009; 31:407–19.
  260. Yan Y, Kluz T, Zhang P, Chen HB, Costa M. Analysis of specific lysine histone H3 and H4 acetylation and methylation status in clones of cells with a gene silenced by nickel exposure. Toxicol Appl Pharmacol, 2003; 190: 272–77.
  261. Yanaihara N, Caplen N, Bowman E et al. Unique MicroRNA molecular profiles in lung cancer diagnosis and prognosis. Toxicol Pathol, 2006; 34:1017–18.
  262. Yanaihara N, Caplen N, Bowman E et al. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell, 2006; 9:189–98.
  263. Yao JY, Zhang L, Zhang X et al. H3K27 trimethylation is an early epigenetic event of p16 (INK4a) silencing for regaining tumorigenesis in fusion reprogrammed hepatoma cells. J Biol Chem, 2010; 285:18828–37.
  264. Yauk C, Polyzos A, Rowan-Carroll A et al. Germ-line mutations, DNA damage, and global hypermethylation in mice exposed to particulate air pollution in an urban/ industrial location. Proc Natl Acad Sci USA 2008; 105: 605–10.
  265. Yuan GC et al. Genome-scale identification of nucleosome positions in S. cerevisiae. Science, 2005; 309:626–630.
  266. Zainuddin N, Kanduri M, Berglund M et al. Quantitative evaluation of p16 (INK4a) promoter methylation using pyrosequencing in de novo diffuse large B-cell lymphoma. Leuk Res, 2011; 35:438–43.
  267. Zeschnigk M, Albrecht B, Buiting K et al. IGF2/H19 hypomethylation in Silver-Russell syndrome and isolated hemihypoplasia. Eur J Hum Genet, 2008;16:328–34.
  268. Zhang A, Feng H, Yang G et al. Unventilated indoor coal-fired stoves in Guizhou province, China: cellular and genetic damage in villagers exposed to arsenic in food and air. Environ Health Perspect, 2007;115:653–8.
  269. Zhang B, Pan X. RDX induces aberrant expression of microRNAs in mouse brain and liver. Environ Health Perspect, 2009;117:231–40.
  270. Zhang C, Jin Y, Xu H et al. Relationship between promoter methylation of p16, DAPK and RAR beta genes and the clinical data of non-small cell lung cancer. Zhonghua Yi Xue Yi Chuan Xue Za Zhi, 2011;28:23–28.
  271. Zhang K, Zhang R, Li X, Yin G, Niu X. Promote r methylation status of p15 and p21 genes in HPP-CFCs of bone marrow of patients with psoriasis. Eur J Dermatol 2009; 19:141–46.
  272. Zhang Y, Wang R, Song H et al. Methylation of multiple genes as a candidate biomarker in non-small cell lung cancer. Cancer Lett, 2011;303:21–8.
  273. Zhang B et al. microRNAs as oncogenes and tumor suppressors. Dev. Biol. 2007;302:1–12.
  274. Zhao CQ, Young MR, Diwan BA, Coogan TP, Waalkes MP. Association of arsenic-induced malignan t transformation with DNA hypomethylation and aberrant gene expression. Proc Natl Acad Sci USA 1997;94: 10907–12.
  275. Zhao F, Chen Y, Zeng L et al. Role of triptolide in cell proliferation, cell cycle arrest, apoptosis and histone methylation in multiple myeloma U266 cells. Eur J Pharmacol, 2010;646:1–11.
  276. Zhao Y, Tian X, Du J et al. Methylation and expression of gene p16INK4a and RB in gastric carcinomas. Beijing Da Xue Xue Bao, 2003;35:382–85.
  277. Zhao YF, Shen SP, Jiang JY, Geng H, Guo JG, Xie LP. Methylation and expression of gene p16INK4a and RB in breast carcinoma. Zhonghua Bing Li Xue Za Zhi 2010; 39: 377–81.
  278. Zhong CX, Mass MJ. Both hypomethylation and hypermethylation of DNA associated with arsenite exposure in cultures of human cells identified by methylationsensitive arbitrarily-primed PCR. Toxicol Lett 2001; 122: 223–34.
  279. Zhou X, Sun H, Ellen TP, Chen H, Costa M. Arsenite alters global histone H3 methylation. Carcinogenesis, 2008; 29:1831–36.
  280. Zhu SM, Si ML, Wu HL, Mo YY. MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem, 2007; 282:14328–36.
  281. Zykova TA, Zhu F, Lu C et al. Lymphokine-activated killer T-cell-originated protein kinase phosphorylation of histone H2AX prevents arsenite-induced apoptosis in RPMI7951 melanoma cells. Clin Cancer Res, 2006; 12: 6884–93.
Source of Financial Support: Nil Conflict of interest: Nil