enviromental chemical exposure and human epigenetics

Upload: fhadla-nisaa

Post on 14-Apr-2018

222 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    1/27

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    2/27

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    3/27

    Table1Effectsofenvironm

    entalchemicalsonepigeneticchanges

    Environmental

    chemicals

    Ep

    igeneticchanges

    In

    vitro/in

    vivo

    Tissue/spe

    cies

    Exampleofd

    iseasespotentially

    associatedw

    iththeobserved

    changesine

    pigeneticchanges

    Arsenic

    DNAmethylation

    Globalhypomethylation

    Invitro

    HumanHaCaTkeratinocytes,80

    humanprostateepithelialcellline

    RWPE-1,8

    1,82

    TRL1

    215ratliver

    epithelialcellline,8

    3

    V79-Cl3

    Chinesehamsterce

    lls226

    Variouscancers227230

    and

    schizophrenia231

    Globalhypomethylation

    Invivo

    129/SvJmice,84

    fisher344Rat,86

    homozygousTg.AC

    mice,87

    gold-

    fish,2

    32

    humanPBL

    233

    Variouscancers227230

    and

    schizophrenia231

    Globalhypomethylation

    andc-Ha-ras

    hypomethylation

    Invivo

    C57BL/6Jmice85

    Variouscancers227230

    and

    schizophrenia231

    Global

    hypermethylation

    Invivo

    HumanPBL88,89

    Colorectalcancer,234236

    renalcell

    carcinoma,23

    7

    acutelymphoblastic

    leukaemia238

    andbladder

    urothelialcellcarcinoma239

    DAP

    Khypermethylation

    Invitro

    Humanuroepithelial

    SV-HUC-1

    cells90

    Variouscancers240251

    P16

    hypermethylation

    Invitro

    Humanmyelomacell

    lineU26691

    Variouscancers241,248,250,252257

    DBC

    1,

    FAM83A,

    ZSCAN12and

    C1QTNF6

    hypermethylation

    Invitro

    HumanUROtsacells92

    Bladdercancer

    ,258

    breastcancer259

    andmaligna

    ntlymphoprolifera-

    tiveneoplasms260

    P53

    hypermethylation

    Invitro

    Humanlungadenoca

    rcinomaA549

    cells93

    Breastcancer261

    and

    hepatoblasto

    ma262

    C-m

    ychypomethylation

    Invitro

    TRL1215ratliverepithelialcells94

    Gastriccancer,263,264

    colon

    cancer,263liv

    ercancer,207,265,266

    kidneycancer207

    andbladder

    cancer267

    C-m

    ycandc-Ha-ras

    hypomethylation

    Invitro

    Syrianhamsterembryocells95

    Gastriccancer,263,264

    colon

    cancer,263liv

    ercancer,207,265,266

    kidneycancer207

    andbladder

    cancer267

    P16

    andRASSF1

    hypermethylation

    Invivo

    A/Jmice96

    Variouscancers241,248,250,252

    257,268,269

    Globalhypomethylation

    andER-alpha

    hypomethylation

    Invivo

    C3Hmice97

    Variouscancers97,227230

    and

    schizophrenia231

    P53

    andP16

    hypermethylation

    Invivo

    HumanPBL98

    Variouscancers241,248,250,252

    257,261,262

    DAP

    Khypermethylation

    Invivo

    Humanbladder,kidn

    eyand

    ureter99

    Variouscancers240251

    (continued)

    ENVIRONMENTAL EPIGENETICS 81

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    4/27

    Table1Continued

    Environmental

    chemicals

    Epigeneticchanges

    In

    vitro/in

    vivo

    Tissue/species

    Exampleofdiseasespotentially

    associatedw

    iththeobserved

    changesinepigeneticchanges

    RASS

    F1AandPRSS3

    hy

    permethylation

    Invivo

    Humanbladder100

    Lungcanceran

    dprostate

    cancer268,269

    P16hypermethylation

    Invivo

    HumanPBL270

    Variouscancers

    241,248,250,252257

    P53hypermethylation

    Invivo

    Humanbasalcellcarc

    inoma102

    Breastcancer26

    1

    and

    hepatoblastom

    a262

    Both

    hypomethylation

    an

    dhypermethyla-

    tio

    nofVHL

    Invitro

    Humankidneycells27

    1

    Renalcellcarcinoma271

    Hist

    onemodification

    #H3

    acetylation

    Invitro

    UROtsaandURO-ASS

    Ccells92

    Renalcellcarcinomas272

    #H4K16acetylation

    Invitro

    UROtsacells104

    Bladdercancer273

    "H3K14acetylation

    Invitro

    NB4cells105

    Diabeticnephro

    pathy274

    "H3S10

    ph

    osphorylation

    "H3

    phosphorylation

    Invitro

    WI-38humandiploid

    fibroblast

    cells106

    Diabeticnephro

    pathy274

    "H3K9acetylation

    Invitro

    HepG2hepatocarcinom

    acells107

    Diabeticnephro

    pathy274

    #H3,H4,H2a,H2b

    ac

    etylation#H3and

    H4methylation

    Invitro

    Drosophilamelanogastertissue

    culturecelllineKC161103

    Heartdisease27

    5

    andtraumatic

    braininjury2

    76

    "H2bmethylation

    "H3K36trimethylation

    Invitro

    Humanlungcarcinom

    aA549

    cells110

    Diabeticnephro

    pathy,274

    multiple

    myeloma277

    andprostate

    cancer278

    #H3K36dimethylation

    "H3K4dimethylation

    "H3K9dimethylation

    Invitro

    Humanlungcarcinom

    aA549

    cells110,279

    Prostatecancer,2

    78

    kidneycancer,278

    lungcancer,2

    80

    HCC281

    and

    AML282

    #H3K27trimethylation

    "H3K4trimethylation

    "H2AXphosphorylation

    Invitro

    RPMI7951melanoma

    cells112

    Ataxiatelangiectasia283

    #H3K18acetylation

    Invitro

    1470.2celllinederivedfrom

    the

    mouseadenocarcino

    maparent

    line284

    Prostatecancer278

    andcolon

    cancer285

    #H3R17methylation

    miR

    NAs

    "miR-222,

    #miR-210

    Invitro

    TK6cellline100

    Variouscancers

    286290

    andAD291

    #miR-19a

    Invitro

    T24cellline115

    Variouscancers

    292300

    (continued)

    82 INTERNATIONAL JOURNAL OF EPIDEMIOLOGY

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    5/27

    Table1Continued

    Environmental

    chemicals

    Epig

    eneticchanges

    In

    vitro/invivo

    Tissue/species

    Exampleofdis

    easespotentially

    associatedwiththeobserved

    changesinepigeneticchanges

    Nickel

    DNA

    methylation

    ATF-1,

    HIF-1,gptandRb

    hyp

    ermethylation

    Invitro

    G12cellline116,117

    Variouscancers3

    01306

    P16hypermethylation

    Invivo

    Mousehistiocytomas119

    Variouscancers2

    41,248,250,252257

    Histo

    nemodification

    "H3K9methylation

    Invitro

    Humanlungcarcinoma

    A549

    cells123,307

    Heartdisease275

    andtraumatic

    braininjury276

    #Aca

    tallfourcore

    hist

    ones

    "H3K9dimethylation

    Invitro

    Humanlungcarcinoma

    A549

    cells,122,124

    G12

    cells,116,123,126,128,2791

    HAEo-cell

    line,120,121

    human(HAE)andrat

    (NRK)cells,125

    Chine

    sehamster

    cellline127

    Lungcancer,308

    heartdisease,2

    75

    chronicglomer

    ulardisease309

    and

    traumaticbraininjury276

    "H2a,H2b

    ubiquitylation

    #H3K4methylation

    #H3K4acetylation

    #H2a,H2b,H3,H4

    acetylation

    #H4K5,H4K8,H4K12,

    H4K

    16acetylation

    Invivo

    Humanlungcarcinoma

    A549

    cells130

    Ataxiatelangiectasia310

    #H2A,H2B,H3,H4

    acetylation(especially

    inH

    2BK12and

    H2B

    K20)

    Invitro

    Humanairwayepithelia

    l1HAEo-

    (HAE)cellline131

    Heartdisease275

    andtraumatic

    braininjury276

    "H3p

    hosphorylation

    Invitro

    Humanlungcarcinoma

    A549

    cells132

    Diabeticnephrop

    athy274

    Cadmium

    DNA

    methylation

    GlobalDNA

    hyp

    omethylation

    Invitro

    K562cell133

    Colorectalcancer

    ,234236

    renalcell

    carcinoma,2

    37

    acutelymphoblastic

    leukaemia,2

    38

    bladderurothelial

    cellcarcinoma239

    InitiallyinducesDNA

    hyp

    omethylation,

    prolongedexposure

    resu

    ltsinDNA

    hyp

    ermethylation

    Invitro

    TRL1215ratlivercells1

    34

    Notapplicable

    miRN

    As

    #miR-146a

    Invivo

    HumanPBL137

    Variouscancers3

    11313

    (continued)

    ENVIRONMENTAL EPIGENETICS 83

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    6/27

    Table1Continued

    Environmental

    chemicals

    Epig

    eneticchanges

    In

    vitro/invivo

    Tissue/species

    Exampleofdis

    easespotentially

    associatedwiththeobserved

    changesinepigeneticchanges

    Chromium

    DNA

    methylation

    P16andhMLH1

    hyp

    ermethylation

    Invivo

    Humanlung143,144

    Variouscancers24

    1,248,250,252257,314316

    Gpthypermethylation

    Invitro

    G12cellline317

    Notapplicable

    Histo

    nemodification

    #H3S-10

    pho

    sphorylation

    Invitro

    Humanlungcarcinoma

    A549

    cells279

    Type2diabetes,2

    74

    heartdisease275

    andtraumatic

    braininjury276

    #H3K4trimethylation

    #H3a

    ndH4acetylation

    "Di

    methylationand

    trim

    ethylationof

    H3K

    9andH3K4

    #H3K27trimethylation

    and

    H3R2

    dim

    ethylation

    Aluminum

    miRN

    As

    "miR-146a

    Invitro

    HNcells149

    AD,3

    18,319

    cardiac

    hypertrophy320

    andvariousca

    ncers321328

    "miR-9,-128,-125b

    Invitro

    HNcells329

    AD,3

    30

    neurodegeneration331

    and

    variouscancers332335

    Mercury

    DNA

    methylation

    Globalhypomethylation

    Invivo

    Braintissuesinpolarb

    ear139

    Neurologicaldiso

    rders336,337

    and

    variouscancer338

    Rnd2hypermethylation

    Invitro

    Mouseembryonicstem

    cells140

    neuronalmigrationdefect339

    Lead

    DNA

    methylation

    Globalhypomethylation

    Invivo

    HumanPBL,1

    41

    newbor

    numbilical

    cordbloodsamples14

    2

    Variouscancers2

    27230

    and

    schizophrenia2

    31

    Pesticides

    DNA

    methylation

    P53hypermethylation

    Invitro

    HumanlungadenocarcinomaA549

    cells93

    Breastcancer261

    and

    hepatoblastoma262

    AlterDNAmethylation

    int

    hegerm

    line

    Invivo

    Rattestis154156

    Potentialeffects

    intheoffspring

    Hypom

    ethylationof

    c-junandc-myc

    Invivo

    Mouseliver158,159

    Gastriccancer,26

    3,264

    colon

    cancer,263

    liver

    cancer,207,265,266

    kidneycancer2

    07

    andbladder

    cancer267

    Globalhypomethylation

    (Alu)

    Invivo

    HumanPBL161,162

    Variouscancers2

    27230

    and

    schizophrenia2

    31

    (continued)

    84 INTERNATIONAL JOURNAL OF EPIDEMIOLOGY

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    7/27

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    8/27

    Table1Continued

    Environmental

    chemicals

    Epigeneticchanges

    In

    vitro/in

    vivo

    Tissue/spec

    ies

    Exampleofdiseasespotentially

    associatedw

    iththeobserved

    changesinepigeneticchanges

    BisphenolA

    DNA

    methylation

    Hypomethylationofthe

    Ag

    outigeneand

    Ca

    bpIAP

    Invivo

    Mouseembryo192

    Micewithhypo

    methylationofthe

    Agoutigeneareobese,diabetic

    andexhibitincreasedcancer

    rates361,362

    Hypomethylationofthe

    ho

    meoboxgene

    Ho

    xa10

    Invivo

    CD-1mice194

    Notapplicable

    Hypermethylationof

    LA

    MP3.

    Invitro

    Breastepithelialcells1

    95

    Breastcancer19

    5

    miR

    NAs

    "miR-146a

    Invitro

    3Aplacentalcells196

    Cardiachypertrophy,320

    AD318,319

    andvariouscancers321328

    Dioxin

    DNA

    methylation

    Igf2hypomethylation

    Invivo

    Ratliver198

    RussellSilversyndrome363365

    and

    variouscance

    rs366370

    AlterationsinDNA

    methylationatmul-

    tip

    legenomicregions

    Invivo

    Splenocyteofmice199

    Notapplicable

    miR

    NAs

    "miR-191

    Invivo

    Ratliver200

    Breastcancer,342

    colorectal

    cancer321,371

    andgastriccancer372

    RDX

    miR

    NAs

    "let-7,miR-15,-16,-26,

    -181#miR-10b

    Invivo

    Mousebrainandliver

    202

    Variouscancers

    325,373380

    "miR-206,-30,-195

    Invivo

    Mousebrainandliver

    202

    Variouscancers

    342,381385

    DES

    miR

    NAs

    #miR-9-3

    Invitro

    Breastepithelialcells2

    05

    Breastcancer20

    5

    Drinking

    water

    DNA

    methylation

    Glob

    alhypomethylation

    c-m

    yc

    hy

    pomethylation

    Invivo

    Miceliver207,208

    Gastriccancer,2

    63,264

    colon

    cancer,263

    livercancer,207,265,266

    kidneycancer207

    andbladder

    cancer267

    PBL,peripheralbloodleucocytes;HCC,hepatocellularcarcinoma;AML,a

    cutemyeloidleukaemia;AD,Alzheimersdisease;HNcells,humanneuralcells;RDX,hexahydro-1,3,5-

    trinitro-1,3,5-triazine;DES,diethylstilbestrol.

    86 INTERNATIONAL JOURNAL OF EPIDEMIOLOGY

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    9/27

    adjacent genes.13,14 Global hypomethylation is asso-ciated with genomic instability and an increased num-ber of mutational events.1518 There are approximately1.4 million Alu repetitive elements (sequences con-taining a recognition site for the restriction enzyme

    AluI)19 and a half a million long interspersed nucleo-tide (LINE-1) elements in the human genome that are

    normally heavily methylated.20 More than one-thirdof DNA methylation occurs in repetitive elements.20

    Because of their high representation throughout thegenome, LINE-1 and Alu have been used as globalsurrogate markers for estimating the genomic DNAmethylation level in cancer tissues,2022 althoughrecent data show lack of correlation with globalmethylation in normal tissues, such as peripheralblood.23 Other types of abnormalities that can beinduced by environmental pollutants are hyper- orhypo-methylation of specific genes or regions, poten-tially associated with aberrant gene transcription.2427

    DNA methylation alterations that directly affectgene expression often occur in the CpG sites located

    in the promoter regions of the genes. Recent evidencehas shown that differentially methylated sites in vari-ous cancer tissues are enriched in sequences, termedCpG island shores, up to 2 kb distant from the tran-scription start site.28 However, to date, gene-specificDNA methylation alterations induced by environmen-tal exposures have been mostly investigated in genepromoter regions. CpG island shores are clearly

    worthy of further investigation in relation to environ-mental exposures, but whether they hold such im-portance in a non-cancer setting remains to bedetermined.

    Histone modifications

    In humans, protection and packaging of the geneticmaterial are largely performed by histone proteins,

    which also offer a mechanism for regulating DNAtranscription, replication and repair.29 Histonesare nuclear globular proteins that can be covalentlymodified by acetylation (Ac), methylation, phosphor-

    ylation, glycosylation, sumoylation, ubiquitinationand adenosine diphosphate (ADP) ribosylation,30,31

    thus influencing chromatin structure and gene ex-pression.32,33 The most common histone modificationsthat have been shown to be modified by environmen-tal chemicals are Ac and methylation of lysine resi-dues in the amino terminal of histone 3 (H3) and H4.

    Histone Ac, with only a single acetyl group added toeach amino acid residue usually, increases gene tran-scriptional activity;3437 whereas histone methylation(Me), found as mono (Me), di-methyl (Me2), andtri-methyl (Me3) group states38 can inhibit or in-crease gene expression depending on the amino acidposition that is modified.3941

    miRNAs

    miRNAs are short single-stranded RNAs of appro-ximately 2024 nucleotides in length that are

    transcribed from DNA but not translated into pro-teins. miRNAs negatively regulate expression oftarget genes at the post-transcriptional level by bind-ing to 30-untranslated regions of target mRNAs.42

    Each mature miRNA is partially complementary tomultiple target mRNAs and directs the RNA-inducedsilencing complex (RISC) to identify the target

    mRNAs for inactivation.43 miRNAs are initially tran-scribed as longer primary transcripts (pri-miRNAs)and processed first by the RNase enzyme complex,and then by Dicer, leading to incorporation of a singlestrand into the RISC. miRNAs guide RISC to interact

    with mRNAs and determine post-transcriptional re-pression. miRNAs are involved in the regulation ofgene expression through the targeting of mRNAsduring cell proliferation, apoptosis, control of stemcell self renewal, differentiation, metabolism, develop-ment and tumour metastasis.44,45 Compared with othermechanisms involved in gene expression, miRNAs actdirectly before protein synthesis and may be more dir-ectly involved in fine-tuning of gene expression or

    quantitative regulation.46,47 Moreover, miRNAs alsoplay key roles in modifying chromatin structure andparticipating in the maintenance of genome stability.48

    miRNAs can regulate various physiological and patho-logical processes, such as cell growth, differentiation,proliferation, apoptosis and metabolism.42,49 More than10 000 miRNAs have been reported in animals, plantsand viruses by using computational and experimentalmethods in miRNA-related public databases. The aber-rant expression of miRNAs has been linked to varioushuman diseases, including Alzheimers disease, cardiachypertrophy, altered heart repolarization, lymphomas,leukaemias, and cancer at several sites.5066

    Environmental pollutants andepigenetic alterationsMetals

    Heavy metals are widespread environmental contam-inants and have been associated with a number ofdiseases, such as cancer, cardiovascular diseases,neurological disorders and autoimmune diseases.67,68

    In recent years, there has been an increasing appreci-ation of the roles of molecular factors in the aetiologyof heavy metal-associated diseases.6971 Several stu-dies showed that metals act as catalysts in the oxida-

    tive deterioration of biological macromolecules.72

    Metal ions induce reactive oxygen species (ROS),and thus lead to the generation of free radicals.72,73

    ROS accumulation can affect epigenetic factors.7479

    Growing data have linked epigenetic alterations withheavy metal exposure.

    Arsenic

    Evidence has been rapidly increasing that exposureto arsenic (As) alters DNA methylation both globallyand in the promoter regions of certain genes.

    ENVIRONMENTAL EPIGENETICS 87

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    10/27

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    11/27

    This finding suggested that gene silencing mediatedby histone deacetylation may play a critical role innickel-induced cell transformation.129 In addition,nickel has also been shown to induce a loss of histonemethylation in vivo and decreased activity of histoneH3K9 demethylase in vitro.123 Nickel also suppresseshistone H4 acetylation in vitro in both yeast and mam-

    malian cells.130,131 Nickel can induce H3 phosphoryl-ation, specifically in serine 10 (H3S10) via activationof the c-jun N-terminal kinase/stress-activated proteinkinase pathway.132

    Cadmium

    Cadmium (Cd) has been shown to alter global DNAmethylation.133 Takiguchi et al.134 demonstrated thatCd inhibits DNMTs and initially induces global DNAhypomethylation in vitro (TRL1215 rat liver cells).However, prolonged exposure was shown to lead toDNA hypermethylation and enhanced DNMTs activityin the same experiment.134 Cd can also decrease

    DNA methylation in proto-oncogenes and promoteoncogenes expression that can result in cellproliferation.133,134

    Transcriptional and post-transcriptional gene regu-lation is critical in responses to Cd exposure, in

    which miRNAs may play an important role.135,136

    Bollati et al.137 have recently demonstrated thatincreased expression of miR-146a in peripheral bloodleucocytes from steel workers was related to inhal-ation of Cd-rich air particles. miRNA-146a expres-sion is regulated by the transcription factornuclear factor-kappa B, which represents an import-ant causal link between inflammation andcarcinogenesis.138

    Other metals

    Mercury (Hg) is widely present in various envir-onmental media and foods at levels that can ad-

    versely affect humans and animals. Exposure to Hghas been associated with brain tissue DNA hypo-methylation in the polar bear.139 Arai et al.140 havestudied the effects of Hg on DNA methylation statusin mouse embryonic stem cells. After 48 or 96 h ofexposure to the chemical, they observed hypermethy-lation of Rnd2 gene in Hg-treated mouse embryonicstem cells.

    Lead is among the most prevalent toxic environmen-tal metals, and has substantial oxidative properties.

    Long-term exposure to lead was shown to alter epi-genetic marks. In the Normative Aging Study, LINE-1methylation levels were examined in association withpatella and tibia lead levels, measured by K-X-Rayfluorescence. Patella lead levels were associated withreduced LINE-1 DNA methylation. The association be-tween lead exposure and LINE-1 DNA methylationmay have implications for the mechanisms of actionof lead on health outcomes, and also suggests thatchanges in DNA methylation may represent a bio-marker of past lead exposure.141 In addition, Pilsner

    et al.142 characterized genomic DNA methylation inthe lower brain stem region from 47 polar bearshunted in central East Greenland between 1999 and2001. They have reported an inverse association be-tween cumulative lead measures and genomic DNAmethylation level.

    Hexavalent chromium [Cr(VI)] is a mutagen and

    carcinogen that has been linked to lung cancer andother adverse health effects in occupational studies.Kondo et al.143 found p16 and hMLH1 hypermethyla-tion in lung cancer patients with past chromateexposure.144 In vitro experiments on cells exposedto binary mixtures of benzo[a]pyrene (B[a]P) andchromium have shown that B[a]P activates Cyp1A1transcriptional responses mediated by the aryl hydro-carbon receptor (AhR), whereas chromium repressesB[a]P-inducible AhR-mediated gene expression145,146

    by inducing cross-links of histone deacetylase 1DNAmethyltransferase 1 (HDAC1DNMT1) complexes tothe Cyp1A1 promoter chromatin and inhibit histonemarks, including phosphorylation of histone H3Ser-10, trimethylation of H3 Lys-4 and various acetyl-ation marks in histones H3 and H4. HDAC1 andDNMT1 inhibitors or depletion of HDAC1 or DNMT1

    with siRNAs blocked the chromium-induced tran-scriptional repression by decreasing the interactionof these proteins with the Cyp1A1 promoter andallowing histone acetylation to proceed. By inhibitingCyp1A1 expression, chromium stimulate the forma-tion of B[a]P DNA adducts. These findings maylink histone modifications to chromium-associateddevelopmental and carcinogenic outcomes.147

    Chromate exposure of human lung A549 cells hasbeen shown to increase the global levels of di- and

    tri-methylated histone H3 lysine 9 (H3K9) and lysine4 (H3K4), but decrease tri-methylated histone H3lysine 27 (H3K27) and di-methylated histone H3 ar-ginine 2 (H3R2). Most interestingly, H3K9 dimethyla-tion was enriched in the human MLH1 gene promoterfollowing chromate exposure, and this was correlated

    with decreased MLH1 mRNA expression. Chromateexposure increased the protein as well as mRNAlevels of G9a, a histone methyltransferase that specif-ically methylates H3K9. This Cr(VI)-induced increasein G9a may account for the global elevation of H3K9dimethylation. Furthermore, supplementation withascorbate, the primary reductant of Cr(VI) and alsoan essential cofactor for the histone demethylase ac-

    tivity, partially reversed the H3K9 dimethylationinduced by chromate. These results suggest thatCr(VI) may target histone methyltransferases anddemethylases, which in turn affect both global andgene promoter-specific histone methylation, leadingto the silencing of specific tumour suppressorgenes.148

    Recent investigations have demonstrated that alu-minum exposure can alter the expression of anumber of miRNAs. miR-146a in human neural cells

    was up-regulated after treatment with aluminium

    ENVIRONMENTAL EPIGENETICS 89

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    12/27

    sulphate. Up-regulation of miR-146a corresponded tothe decreased expression of complement factor H, arepressor of inflammation.149 In addition, a study onaluminium-sulphate-treated human neural cells inprimary culture has shown increased expression of aset of miRNAs, including miR-9, miR-125b andmiR-128.150 The same miRNAs were also found to

    be up-regulated in brain cells of Alzheimer patients,suggesting that aluminum exposure may induce gen-otoxicity via miRNA-related regulatory elements.150

    Pesticides

    Growing evidence suggests that epigenetic eventscan be induced by pesticide exposures.28,151153

    Animal models have shown that exposure to somepesticides, such as vinclozolin and methoxyclor, in-duces heritable alterations of DNA methylationin male germline associated with testis dysfunc-tion,154156 or affects ovarian function via alteredmethylation patterns.157 Decreased methylation in

    the promoter regions of c-jun and c-myc and increasedlevels of their mRNAs and proteins were foundin livers of mice exposed to dichloro- and trichloro-acetic acid.158,159 Dichlorvos has been demonstratedto induce DNA methylation in multiple tissues inan animal toxicity study.160 DNA methylation in re-petitive elements in blood DNA was inversely asso-ciated with increased levels of plasma pesticideresidues and other persistent organic pollutantsin an Arctic population,161 a finding later confirmedin a similar study in a Korean population.162 Whetheraberrant DNA methylation represents the link be-tween pesticides and risks of pesticide-related disease,including the excess of cancer risk observed in

    some epidemiology studies,

    163168

    remains to bedetermined.Dieldrin, a widely used organochlorine pesticide, has

    been shown to increase acetylation of core histonesH3 and H4 in a time-dependent manner. Histoneacetylation was induced within 10 min of dieldrin ex-posure, suggesting that histone hyperacetylation is anearly event in dieldrin-induced diseases. Treatment

    with anacardic acid, a histone acetyltransferaseinhibitor, decreased dieldrin-induced histone acetyl-ation.169 Dieldrin was further shown to induce his-tone hyperacetylation in the striatum and substantianigra in mouse models, suggesting the roles forhistone hyperacetylation in dieldrin-induced dopa-

    minergic neuronal degeneration.170

    Air pollution

    Exposure to particulate matter (PM) of ambient airpollution has been associated with increased morbid-ity and mortality related to cardiovascular and re-spiratory diseases.171,172 Black carbon, a componentof PM derived from vehicular traffic, has beenlinked to decreased DNA methylation in LINE-1 re-petitive elements in 1097 blood DNA samples of

    elderly men in the Boston area. Additional evidencefor PM effects on DNA methylation stemmed from aninvestigation of workers in a steel plant with

    well-characterized exposure to PM with diameters of

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    13/27

    poly (ADP-ribose) polymerases-1 (PARP-1), a geneinvolved in DNA repair.188

    Bisphenol A

    Bisphenol A (BPA) is an endocrine disruptor withpotential reproductive effects, as well as a weakcarcinogen associated with increased cancer risk inadult life through fetal exposures.189,190 BPA is

    widely used as an industrial plasticizer in epoxyresins for food and beverage containers, baby bottlesand dental composites.191 Dolinoy et al.192 reportedthat periconceptional exposure to BPA shiftedthe coat colour distribution of the viable yellowagouti (Avy) mouse offspring toward yellow bydecreasing CpG methylation in an intracisternal Aparticle (IAP) retrotransposon upstream of the

    Agouti gene.193 In this animal model, the yellow-coat phenotype is associated with increased cancerrates, as well as with obesity and insulin resistance.In the same set of experiments, maternal dietary sup-

    plementation, with either methyl donors like folicacid or the phytoestrogen genistein, blunted theeffect of BPA on IAP methylation and prevented thecoat colour change caused by BPA exposure.192 Inpregnant CD-1 mice treated with BPA, Bromeret al.194 found decreased methylation and increasedexpression of the homeobox gene Hoxa10, which con-trols uterine organogenesis. In breast epithelial cellstreated with low-dose BPA, gene expression profilingidentified 170 genes with expression changes in re-sponse to BPA, of which expression of lysosomal-associated membrane protein 3 (LAMP3) was shownto be silenced due to DNA hypermethylation in itspromoter.195

    In a recent study by Avissar-Whiting et al.,

    196

    anelevated expression of miR-146a was observed inBPA-treated placental cell lines and miR-146a expres-sion was associated with slower cell proliferationand higher sensitivity to the bleomycin-inducedDNA damage.

    Dioxin

    Dioxin is a compound that has been classified as ahuman carcinogen by the International Agency forResearch on Cancer. As dioxin is only a weak muta-gen, extensive research has been conducted to identifypotential mechanisms contributing to carcinogenesis.One proposed pathway to carcinogenesis is related to

    the powerful dioxin-induced activation of microsomalenzymes, such as CYP1B1, that might activateother procarcinogen compounds to active carcinogen.The capability of dioxin to induce CYP1B1 has beenrecently shown in vitro to depend on the methylationstate of the CYP1B1 promoter.197 Also, dioxin wasshown to reduce the DNA methylation level of Igf2in rat liver.198 Recently, alterations in DNA methyla-tion at multiple genomic regions were identified insplenocytes of mice treated with dioxin, a finding

    potentially related to dioxin immunotoxicity.199 In axenograft mouse model of hepatocellular carcinoma,Elyakim et al.200 have also found that dioxinup-regulated miR-191. In the same study, inhibitionof miR-191 inhibited apoptosis and decreased cell pro-liferation, suggesting that increased miR-191 expres-sion may contribute to determine dioxin-induced

    carcinogenicity.

    Hexahydro-1,3,5-trinitro-1,3,5-triazine(RDX, also known as hexogen or cyclonite)

    Hexahydro-1,3,5-trinitro-1,3,5-triazine (commonlyknown as RDX, the British code name for RoyalDemolition Explosive) is an explosive polynitramineand common ammunition constituent used in mili-tary and civil activities. Although most of this envir-onmental pollutant is found in soils, RDX and itsmetabolites are also found in water sources.201

    Exposure to RDX and its metabolites could causeneurotoxicity, immunotoxicity and cancers.202 Zhang

    et al.202

    have recently evaluated the effects of RDX onmiRNA expression in mouse brain and liver. In thisstudy, out of 113 miRNAs, 10 were up-regulated and3 were down-regulated. Most of the miRNAs thatshowed altered expression, including let-7, miR-17-92, miR-10b, miR-15, miR-16, miR-26 and miR-181, were found to regulate toxicant-metabolizingenzymes, as well as genes related to carcinogenesisand neurotoxicity.202

    Diethylstilbestrol

    Diethylstilbestrol (DES) is a synthetic oestrogen thatwas used to prevent miscarriages in pregnant womenbetween the 1940s and the 1960s.203 A moderate in-crease in breast cancer risk has been shown both indaughters of women who were treated with DESduring pregnancy, as well as in their daughters.204

    Hsu et al.205 have demonstrated that the expressionof 82 miRNAs (9.1% of the 898 miRNAs evaluated)

    were altered in breast epithelial cells when exposed toDES. In particular, the suppression of miR-9-3 expres-sion was accompanied by promoter hypermethylationof the miR-9-3 coding gene in DES-treated epithelialcells.205

    Chemicals in drinking water

    Chlorination by-products are formed as a result of the

    water chlorination for anti-fouling purposes. Variouschlorination by-products in drinking water, such astriethyltin,206 chloroform207 and trihalomethanes,208

    have been questioned for potential adverse healtheffects.209 These chemicals have been shown toinduce certain epigenetic changes. Rats that werechronically intoxicated with triethyltin in drinking

    water showed development of cerebral oedema aswell as an increase of phosphatidylethanolamine-N-methyltransferase activities. This increased

    ENVIRONMENTAL EPIGENETICS 91

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    14/27

    methylation might be a compensatory mechanism forcounteracting the membrane damages induced bytriethyltin.206 Chloroform, dichloroacetic acid (DCA)and trichloroacetic acid (TCA), three liver and kidneycarcinogens, are by-products of chlorine disinfectionfound in drinking water.210,211 Mice treated withDCA, TCA and chloroform show global hypomethyla-

    tion and increased expression of c-myc, a proto-oncogene involved in liver and kidney tumours.207

    Trihalomethanes (chloroform, bromodichloro-methane, chlorodibromomethane and bromoform) areregulated organic contaminants in chlorinated drink-ing water. In female B6C3F1 mouse liver, trihalo-methanes demonstrated carcinogenic activity.Chloroform and bromodichloromethane decreased thelevel of 5-methylcytosine in hepatic DNA. Methylationin the promoter region of the c-myc gene was reducedby the trihalomethanes, consistent with their carcino-genic activity.208

    Environmental epigenomics:challenges and opportunitiesfor epidemiologic studiesThe studies reviewed in this article have demonstratedthe potential effects of environmental pollutants onthe epigenome. Several of the epigenomic changesobserved in response to environmental exposuresmight be mechanistically associated with susceptibil-ity to diseases (Table 1). Further studies of epigeneticmechanisms in disease pathogenesis, including therole of epigenetics in the developmental origins ofhealth and disease, their relationships with environ-

    mental exposures and the pathways associated withthe disease phenotype may help develop preventiveand therapeutic strategies.

    Epigenetics and developmental originsof health and disease

    During embryogenesis, epigenetic patterns changedynamically to adapt embryos to be fit for furtherdifferentiation.7 Two waves of epigenetic reprogram-ming, which take place at the zygote stage and duringprimordial germ cells formation, accompany mamma-lian development.212

    Experiments on mice carrying the Avy have demon-strated that embryo life is a window of exquisite sen-

    sitivity to the environment. In viable yellow (Avy/a)mice, transcription originating in a IAP retrotrans-poson inserted upstream of the agouti gene (A)causes ectopic expression of agouti protein, result-ing in yellow fur, obesity, diabetes and increasedsusceptibility to tumours.213 BPA is a high-production-volume chemical used in the manufactureof polycarbonate plastic. In utero or neonatal expos-ure to BPA is associated with higher body weight,increased breast and prostate cancer and alteredreproductive function.

    Additional experimental studies have suggested epi-genetic mechanisms as potential intermediates forthe effects of prenatal exposures to pesticides suchas vinclozolin and methoxyclor,154 as well as of other conditions such as nutritional supplies ofmethyl donors.192 Evidence has also been accumulat-ing in humans. Investigations of candidate loci among

    individuals prenatally exposed to poor nutritionduring the Dutch famine in 194445 indicate that epi-genetic changes induced by prenatal exposures maybe common in humans, although they appear to berelatively small and greatly dependent on the timingof the exposure during gestation.214,215 Based on find-ings of changes in DNA methylation in subjectsexposed to the Dutch famine, Heijmans et al.216

    have suggested that the epigenome may representa molecular archive of the prenatal environment,

    via which the in-utero environment may produce ser-ious ramifications on health and disease later in life.Terry et al.217 found that prenatal exposure to cigar-ette smoke was associated with increased overallblood DNA methylation level in adulthood. Otherexamples include decreased LINE-1 and Sat 2 methy-lation level in adults and children prenatally exposedto smoking,218 and global DNA hypomethyla-tion in newborns with utero exposures of maternalsmoking.219 In addition to these DNA methylationchanges, Maccani et al.220 have recently observedthat miR-16, miR-21 and miR-146a were down-regulated in cigarette smoke-exposed placentas com-pared to controls.

    Additional well-conducted epigenetic studies arenow warranted to generate a catalogue of regionsthat are sensitive to the prenatal environment and

    may reflect developmental influences on humandisease.

    Can we develop epigenomic biosensors ofpast exposures?

    An important property of epigenomic signatures isthat, because they can be propagated through cell div-ision even in cells with high turnover, they can persisteven after the exposure is removed. In addition, asdiscussed above, an individuals epigenome may alsoreflect his/her prenatal environmental exposure ex-perience. Thus, epigenomic profiling of individualsexposed to environmental pollutants might provide

    biosensors or molecular archives of ones past oreven prenatal environmental exposures. Using epige-nomics, exposure assessment might be brought to re-search investigations and preventive settings whererepeated collections of exposure data might be un-feasible or exceedingly expensive. Further research isneeded to establish how rapid are the changesinduced by environmental pollutants, as well as

    whether they accumulate in response to repeated orcontinuous exposure and how long they persist afterthe exposure is removed.

    92 INTERNATIONAL JOURNAL OF EPIDEMIOLOGY

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    15/27

    What are suitable study designs andapproaches for environmental epigenomics?

    The field of environmental epigenetics has evolvedrapidly in the past several years. As research applica-tions grow, investigators will be facing several diffi-culties and challenges. Some studies have producedinconsistent results on same pollutants. Several fac-

    tors may contribute to the inconsistencies. Epigeneticalterations are tissue specific.221 It is conceivable thatthe same environmental pollutant may produce differ-ent epigenetic changes in different tissues, and even

    within the same tissue on different cell types. Largerstudies with well-defined exposure information thatallows examining epigenetic changes across differenttissues are needed. Different study design, small sam-ple size and different laboratory methods may also bemajor causes for the inconsistency. Replicating resultsand identifying the sources of variability across stu-dies is a major challenge for epigenetic investigations.Because epigenetic markers change over time, diseaseoutcomes are prone to reverse causation, i.e. an asso-

    ciation between a disease and an epigenetic markermay be determined by an influence of the disease onthe epigenetic patterns, rather than vice versa.222

    Although epigenetic alterations that were found tobe induced by or associated with environmental pol-lutants were also found in various diseases, almost nostudy has examined the sequence of exposures, epi-genetic alterations and diseases.

    Longitudinal studies with prospective collection ofobjective measures of exposure, biospecimens for epi-genetic analyses and preclinical and clinical diseaseoutcomes are needed to appropriately establish caus-ality. Existing prospective epidemiology investigationsmight provide resources for mapping epigenomic

    changes in response to specific chemicals. However,cohort studies in which biospecimens have been pre-

    viously collected for genetic or biochemical studiesmight pose several challenges. Most studies have col-lected biospecimens, such as blood, urine or buccalcells, which might not necessarily participate in theaetiology of the disease of interest. Methods of collec-tion and processing (e.g. whole blood vs buffy coat)might modify the cell types stored, thus potentiallyimpacting on epigenetic marks. In addition, high-coverage methods providing high-dimensional dataon DNA methylation, histone modifications andmiRNA expression are increasingly used in humaninvestigations.

    Albeit epigenetic mechanisms have propertiesthat make them ideal molecular intermediates of en-

    vironmental effects, the proportion of the effects ofany individual environmental exposure that mightbe mediated through epigenetic mechanisms isstill undetermined. Epidemiology and statisticalapproaches, including well-designed prospectivestudies and advanced statistical methods for causalinference are urgently needed. Similarly to genomicstudies,223 epidemiological causal reasoning in

    epigenomics should include careful consideration ofknowledge, data, methods and techniques from mul-tiple disciplines.

    The potential interactions between differentforms of epigenetic modification

    Most studies in environmental epigenetics have sep-arately evaluated only one of the types of the epigen-etic marks, i.e. DNA methylation, histonemodifications or miRNA expression. However, epigen-etic marks are related by an intricate series of inter-actions that may generate a self-reinforcing cycle ofepigenetic events directed to control gene expres-sion.224 For instance, histone deacetylation andmethylation at specific amino acid residues con-tribute to the establishment of DNA methylation pat-terns. miRNA expression is controlled by DNAmethylation in miRNA encoding genes, and, in turn,miRNAs have been shown to modify DNA methyla-tion.225 Future studies that include comprehensive

    investigations of multiple epigenetic mechanismsmight help elucidate the timing and participation ofDNA methylation, histone modifications and miRNAsto determine environmental effects on diseasedevelopment.

    Can epigenomics be used for prevention?

    One major objective of epidemiology investigations isto provide the groundwork for future preventive inter-

    ventions. Numerous clinical and preclinical studiesshowed that most of the epigenetic changes are re-

    versible, which offers novel insights to develop newpreventive and therapeutic strategies that might takeadvantage of molecules that modify the activities ofepigenetic enzymes, such as DNMTs and HDACs, as

    well as of the growing field of RNAi therapeutics.Drugs have been designed and developed that pro-duce functional effects, such as histone acetylationand DNA hypomethylation that might be used to re-store the normal transcription level of genes. Futureepidemiology studies have a unique opportunity toevaluate whether the effects of environmental expos-ures on the epigenome are mitigated by positivechanges in lifestyles, or worsened by the interaction

    with other risk factors. Future epigenomic researchmay provide information for developing preventivestrategies, including exposure reduction, as well aspharmacological, dietary or lifestyle interventions.

    FundingOur work is partially supported by grants from theHSPH-NIEHS Center for Environmental Health NewInvestigator Fund (P30ES000002) and NIH award1RC1ES018461-01.

    Conflict of interest: None declared.

    ENVIRONMENTAL EPIGENETICS 93

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    16/27

    KEY MESSAGES

    Rapidly growing evidence has linked environmental pollutants with epigenetic variations, includingchanges in DNA methylation, histone modifications and microRNAs.

    Some of such epigenetic changes have been associated with various diseases.

    Further studies of epigenetic mechanisms in disease pathogenesis, their relationships with environ-mental exposures and related pathways are needed for the development of preventive and therapeuticstrategies.

    Future epidemiology studies on environmental pollutants and epigenome face several challenges.

    References1 Pruss-Ustun Annette CC. Preventing disease through

    healthy environments. Towards an estimate of the envir-onmental burden of disease World Health Organization(WHO), 2006.

    2 Bollati V, Baccarelli A. Environmental epigenetics. Heredity2010;105:10512.

    3 Bezek S, Ujhazy E, Mach M, Navarova J, Dubovicky M.

    Developmental origin of chronic diseases: toxicologicalimplication. Interdiscip Toxicol 2008;1:2931.

    4 Tang WY, Ho SM. Epigenetic reprogramming and im-printing in origins of disease. Rev Endocr Metab Disord2007;8:17382.

    5 Vaissiere T, Sawan C, Herceg Z. Epigenetic interplay be-tween histone modifications and DNA methylation ingene silencing. Mutat Res 2008;659:4048.

    6 Grewal SI, Moazed D. Heterochromatin and epi-genetic control of gene expression. Science 2003;301:798802.

    7 Reik W, Dean W, Walter J. Epigenetic reprogramm-ing in mammalian development. Science 2001;293:108993.

    8 Baccarelli A, Bollati V. Epigenetics and environmental

    chemicals. Curr Opin Pediatr 2009;21:24351.9 Heightman TD. Therapeutic prospects for epigenetic

    modulation. Expert Opin Ther Targets 2011;15:72940.10 Wright RJ. Epidemiology of stress and asthma: from con-

    stricting communities and fragile families to epigenetics.Immunol Allergy Clin North Am 2011;31:1939.

    11 Miller OJ, Schnedl W, Allen J, Erlanger BF.5-Methylcytosine localised in mammalian constitutiveheterochromatin. Nature 1974;251:63637.

    12 Yoder JA, Walsh CP, Bestor TH. Cytosine methylation andthe ecology of intragenomic parasites. Trends Genet 1997;13:33540.

    13 Jones PA, Baylin SB. The fundamental role of epigeneticevents in cancer. Nat Rev Genet 2002;3:41528.

    14 Bird A. DNA methylation patterns and epigenetic

    memory. Genes Dev 2002;16:621.15 Chen RZ, Pettersson U, Beard C, Jackson-Grusby L,

    Jaenisch R. DNA hypomethylation leads to elevated mu-tation rates. Nature 1998;395:8993.

    16 Eden A, Gaudet F, Waghmare A, Jaenisch R.Chromosomal instability and tumors promoted by DNAhypomethylation. Science 2003;300:455.

    17 Gaudet F, Hodgson JG, Eden A et al. Induction of tumorsin mice by genomic hypomethylation. Science 2003;300:48992.

    18 Laird PW. Cancer epigenetics. Hum Mol Genet 2005;(Spec No 1):R6576.

    19 Houck CM, Rinehart FP, Schmid CW. A ubiquitous familyof repeated DNA sequences in the human genome. J MolBiol 1979;132:289306.

    20 Yang AS, Estecio MR, Doshi K, Kondo Y, Tajara EH,Issa JP. A simple method for estimating global DNAmethylation using bisulfite PCR of repetitive DNA elem-ents. Nucleic Acids Res 2004;32:e38.

    21 Weisenberger DJ, Campan M, Long TI et al. Analysis ofrepetitive element DNA methylation by MethyLight.Nucleic Acids Res 2005;33:682336.

    22 Zhu ZZ, Hou L, Bollati V et al. Predictors of global methy-lation levels in blood DNA of healthy subjects: a com-bined analysis. Int J Epidemiol 2012;41:12639.

    23 Choi JY, James SR, Link PA et al. Association betweenglobal DNA hypomethylation in leukocytes and risk ofbreast cancer. Carcinogenesis 2009;30:188997.

    24 Gronniger E, Weber B, Heil O et al. Aging and chronic sunexposure cause distinct epigenetic changes in humanskin. PLoS Genet 2010;6:e1000971.

    25 Deshmukh RS, Ostrup O, Ostrup E et al. DNA methylationin porcine preimplantation embryos developed in vivoand produced by in vitro fertilization, parthenogeneticactivation and somatic cell nuclear transfer. Epigenetics2011;6:17787.

    26 Anier K, Malinovskaja K, Aonurm-Helm A, Zharkovsky A,Kalda A. DNA methylation regulates cocaine-induced be-havioral sensitization in mice. Neuropsychopharmacology2010;35:245061.

    27 Orta ML, Dominguez I, Pastor N, Cortes F, Mateos S. Therole of the DNA hypermethylating agent Budesonide inthe decatenating activity of DNA topoisomerase II. MutatRes 2010;694:4552.

    28 Irizarry RA, Ladd-Acosta C, Wen B et al. The human coloncancer methylome shows similar hypo- and hypermethy-lation at conserved tissue-specific CpG island shores. NatGenet 2009;41:17886.

    29 Shahbazian MD, Grunstein M. Functions of site-specifichistone acetylation and deacetylation. Annu Rev Biochem2007;76:75100.

    30 Suganuma T, Workman JL. Crosstalk among HistoneModifications. Cell 2008;135:60407.

    31 Zheng YG, Wu J, Chen Z, Goodman M. Chemical regula-tion of epigenetic modifications: opportunities for newcancer therapy. Med Res Rev 2008;28:64587.

    32 Kouzarides T. Chromatin modifications and their func-tion. Cell 2007;128:693705.

    33 Luger K, Mader AW, Richmond RK, Sargent DF,Richmond TJ. Crystal structure of the nucleosome coreparticle at 2.8 A resolution. Nature 1997;389:25160.

    34 Glozak MA, Seto E. Histone deacetylases and cancer.Oncogene 2007;26:542032.

    94 INTERNATIONAL JOURNAL OF EPIDEMIOLOGY

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    17/27

    35 Sterner DE, Berger SL. Acetylation of histones andtranscription-related factors. Microbiol Mol Biol Rev 2000;64:43559.

    36 Cress WD, Seto E. Histone deacetylases, transcriptionalcontrol, and cancer. J Cell Physiol 2000;184:116.

    37 Wang Z, Zang C, Rosenfeld JA et al. Combinatorial pat-terns of histone acetylations and methylations in thehuman genome. Nat Genet 2008;40:897903.

    38 Klose RJ, Zhang Y. Regulation of histone methylation bydemethylimination and demethylation. Nat Rev Mol CellBiol 2007;8:30718.

    39 Martin C, Zhang Y. The diverse functions of histonelysine methylation. Nat Rev Mol Cell Biol 2005;6:83849.

    40 Wysocka J, Allis CD, Coonrod S. Histone arginine methy-lation and its dynamic regulation. Front Biosci 2006;11:34455.

    41 Meissner A, Mikkelsen TS, Gu H et al. Genome-scale DNAmethylation maps of pluripotent and differentiated cells.Nature 2008;454:76670.

    42 Singh SK, Pal Bhadra M, Girschick HJ, Bhadra U.MicroRNAsmicro in size but macro in function. FEBS J2008;275:492944.

    43 Matkovich SJ, Van Booven DJ, Eschenbacher WH,

    Dorn GW 2nd. RISC RNA sequencing for context-specificidentification of in vivo microrna targets. Circ Res 2010;108:1826.

    44 Williams AE. Functional aspects of animal microRNAs.Cell Mol Life Sci 2008;65:54562.

    45 Huang Y, Shen XJ, Zou Q, Wang SP, Tang SM, Zhang GZ.Biological functions of microRNAs: a review. J PhysiolBiochem 2011;67:12939.

    46 Ying SY, Chang DC, Lin SL. The microRNA (miRNA):overview of the RNA genes that modulate gene function.Mol Biotechnol 2008;38:25768.

    47 Bartel DP, Chen CZ. Micromanagers of gene expression:the potentially widespread influence of metazoanmicroRNAs. Nat Rev Genet 2004;5:396400.

    48 Guil S, Esteller M. DNA methylomes, histone codes and

    miRNAs: tying it all together. Int J Biochem Cell Biol 2009;41:8795.

    49 Backes C, Meese E, Lenhof HP, Keller A. A dictionary onmicroRNAs and their putative target pathways. NucleicAcids Res 2010;38:447686.

    50 Ho L, Fivecoat H, Wang J, Pasinetti GM. Alzheimers dis-ease biomarker discovery in symptomatic and asymptom-atic patients: experimental approaches and future clinicalapplications. Exp Gerontol 2010;45:1522.

    51 Provost P. Interpretation and applicability of microRNAdata to the context of Alzheimers and age-related dis-eases. Aging 2010;2:16669.

    52 Provost P. MicroRNAs as a molecular basis for mentalretardation, Alzheimers and prion diseases. Brain Res2010;1338:5866.

    53 Cheng Y, Zhang C. MicroRNA-21 in cardiovascular dis-ease. J Cardiovasc Transl Res 2010;3:25155.

    54 Montgomery RL, van Rooij E. MicroRNA regulation as atherapeutic strategy for cardiovascular disease. Curr DrugTargets 2010;11:93642.

    55 Shen E, Diao X, Wei C, Wu Z, Zhang L, Hu B. MicroRNAstarget gene and signaling pathway by bioinformatics ana-lysis in the cardiac hypertrophy. Biochem Biophys ResCommun 2010;397:38085.

    56 Swynghedauw B, Delcayre C, Samuel JL, Mebazaa A,Cohen-Solal A. Molecular mechanisms in evolutionarycardiology failure. Ann N Y Acad Sci 2010;1188:5867.

    57 Fabbri M, Croce CM, Calin GA. MicroRNAs in the on-togeny of leukemias and lymphomas. Leuk Lymphoma2009;50:16070.

    58 Garzon R, Croce CM. MicroRNAs in normal and malig-nant hematopoiesis. Curr Opin Hematol 2008;15:35258.

    59 Olive V, Jiang I, He L. mir-17-92, a cluster of miRNAs inthe midst of the cancer network. Int J Biochem Cell Biol2010;42:134854.

    60 Marcucci G, Radmacher MD, Mrozek K, Bloomfield CD.MicroRNA expression in acute myeloid leukemia. CurrHematol Malig Rep 2009;4:8388.

    61 Motyckova G, Stone RM. The role of molecular tests inacute myelogenous leukemia treatment decisions. CurrHematol Malig Rep 2010;5:10917.

    62 Zhao H, Wang D, Du W, Gu D, Yang R. MicroRNA andleukemia: tiny molecule, great function. Crit Rev OncolHematol 2010;74:14955.

    63 Chen J, Xu X. Diet, epigenetic, and cancer prevention. AdvGenet 2010;71:23755.

    64 Garzon R, Marcucci G, Croce CM. Targeting microRNAsin cancer: rationale, strategies and challenges. Nat RevDrug Discov 2010;9:77589.

    65 Lin PY, Yu SL, Yang PC. MicroRNA in lung cancer. Br J

    Cancer 2010;103:114448.66 Mathers JC, Strathdee G, Relton CL. Induction of epigen-

    etic alterations by dietary and other environmental fac-tors. Adv Genet 2010;71:339.

    67 Howard Hu. Human health and heavy metals exposure.In: McCally M (ed.). Life Support: The Environment andHuman Health. Boston: Massachusetts Institute ofTechnology, 2002.

    68 Hemdan NY, Emmrich F, Faber S, Lehmann J, Sack U.Alterations of TH1/TH2 reactivity by heavy metals: pos-sible consequences include induction of autoimmune dis-eases. Ann NY Acad Sci 2007;1109:12937.

    69 Waalkes MP. Cadmium carcinogenesis. Mutat Res 2003;533:10720.

    70 Salnikow K, Zhitkovich A. Genetic and epigenetic mech-

    anisms in metal carcinogenesis and cocarcinogenesis:nickel, arsenic, and chromium. Chem Res Toxicol 2008;21:2844.

    71 Huang C, Ke Q, Costa M, Shi X. Molecular mechanismsof arsenic carcinogenesis. Mol Cell Biochem 2004;255:5766.

    72 Galaris D, Evangelou A. The role of oxidative stress inmechanisms of metal-induced carcinogenesis. Crit RevOncol Hematol 2002;42:93103.

    73 Leonard SS, Bower JJ, Shi X. Metal-induced toxicity, car-cinogenesis, mechanisms and cellular responses. Mol CellBiochem 2004;255:310.

    74 Monks TJ, Xie R, Tikoo K, Lau SS. Ros-induced histonemodifications and their role in cell survival and celldeath. Drug Metab Rev 2006;38:75567.

    75 Donaldson K, Stone V, Borm PJ et al. Oxidative stressand calcium signaling in the adverse effects of environ-mental particles (PM10). Free Radic Biol Med 2003;34:136982.

    76 Gilmour PS, Rahman I, Donaldson K, MacNee W. Histoneacetylation regulates epithelial IL-8 release mediated byoxidative stress from environmental particles. Am JPhysiol Lung Cell Mol Physiol 2003;284:L53340.

    77 Babar IA, Slack FJ, Weidhaas JB. miRNA modulation ofthe cellular stress response. Future Oncol 2008;4:28998.

    78 Sakano K, Inagaki Y, Oikawa S, Hiraku Y, Kawanishi S.Copper-mediated oxidative DNA damage induced by

    ENVIRONMENTAL EPIGENETICS 95

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    18/27

    eugenol: possible involvement of O-demethylation. MutatRes 2004;565:3544.

    79 Galaris D, Skiada V, Barbouti A. Redox signaling andcancer: the role of labile iron. Cancer Lett 2008;266:2129.

    80 Reichard JF, Schnekenburger M, Puga A. Long termlow-dose arsenic exposure induces loss of DNA methyla-tion. Biochem Biophys Res Commun 2007;352:18892.

    81 Benbrahim-Tallaa L, Waterland RA, Styblo M,Achanzar WE, Webber MM, Waalkes MP. Molecularevents associated with arsenic-induced malignant trans-formation of human prostatic epithelial cells: aberrantgenomic DNA methylation and K-ras oncogene activa-tion. Toxicol Appl Pharmacol 2005;206:28898.

    82 Coppin JF, Qu W, Waalkes MP. Interplay between cellu-lar methyl metabolism and adaptive efflux during onco-genic transformation from chronic arsenic exposure inhuman cells. J Biol Chem 2008;283:1934250.

    83 Zhao CQ, Young MR, Diwan BA, Coogan TP,Waalkes MP. Association of arsenic-induced malignanttransformation with DNA hypomethylation and aber-rant gene expression. Proc Natl Acad Sci USA 1997;94:1090712.

    84 Chen H, Li S, Liu J, Diwan BA, Barrett JC, Waalkes MP.Chronic inorganic arsenic exposure induces hepatic globaland individual gene hypomethylation: implications for ar-senic hepatocarcinogenesis. Carcinogenesis 2004;25:177986.

    85 Okoji RS, Yu RC, Maronpot RR, Froines JR. Sodiumarsenite administration via drinking water increasesgenome-wide and Ha-ras DNA hypomethylation inmethyl-deficient C57BL/6J mice. Carcinogenesis 2002;23:77785.

    86 Uthus EO, Davis C. Dietary arsenic affectsdimethylhydrazine-induced aberrant crypt formationand hepatic global DNA methylation and DNA methyl-transferase activity in rats. Biol Trace Elem Res 2005;103:13345.

    87 Xie Y, Trouba KJ, Liu J, Waalkes MP, Germolec DR.Biokinetics and subchronic toxic effects of oral arsenite,arsenate, monomethylarsonic acid, and dimethylarsinicacid in v-Ha-ras transgenic (Tg.AC) mice. Environ HealthPerspect 2004;112:125563.

    88 Majumdar S, Chanda S, Ganguli B, Mazumder DN,Lahiri S, Dasgupta UB. Arsenic exposure induces genomichypermethylation. Environ Toxicol 2010;25:31518.

    89 Pilsner JR, Liu X, Ahsan H et al. Genomic methylation ofperipheral blood leukocyte DNA: influences of arsenicand folate in Bangladeshi adults. Am J Clin Nutr 2007;86:117986.

    90 Chai CY, Huang YC, Hung WC, Kang WY, Chen WT.Arsenic salts induced autophagic cell death and hyper-methylation of DAPK promoter in SV-40 immortalized

    human uroepithelial cells. Toxicol Lett 2007;173:4856.91 Fu HY, Shen JZ. Hypermethylation of CpG island of p16

    gene and arsenic trioxide induced p16 gene demethyla-tion in multiple myeloma. Zhonghua Nei Ke Za Zhi 2005;44:41114.

    92 Jensen TJ, Novak P, Eblin KE, Gandolfi AJ, Futscher BW.Epigenetic remodeling during arsenical-induced malig-nant transformation. Carcinogenesis 2008;29:1500508.

    93 Mass MJ, Wang L. Arsenic alters cytosine methylationpatterns of the promoter of the tumor suppressor genep53 in human lung cells: a model for a mechanism ofcarcinogenesis. Mutat Res 1997;386:26377.

    94 Chen H, Liu J, Zhao CQ, Diwan BA, Merrick BA,Waalkes MP. Association of c-myc overexpres-sion and hyperproliferation with arsenite-induced malig-nant transformation. Toxicol Appl Pharmacol 2001;175:26068.

    95 Takahashi M, Barrett JC, Tsutsui T. Transformation byinorganic arsenic compounds of normal Syrian hamsterembryo cells into a neoplastic state in which theybecome anchorage-independent and cause tumors innewborn hamsters. Int J Cancer 2002;99:62934.

    96 Cui X, Wakai T, Shirai Y, Hatakeyama K, Hirano S.Chronic oral exposure to inorganic arsenate interfereswith methylation status of p16INK4a and RASSF1Aand induces lung cancer in A/J mice. Toxicol Sci 2006;91:37281.

    97 Waalkes MP, Liu J, Chen H et al. Estrogen signaling inlivers of male mice with hepatocellular carcinomainduced by exposure to arsenic in utero. J Natl CancerInst 2004;96:46674.

    98 Chanda S, Dasgupta UB, Guhamazumder D et al. DNAhypermethylation of promoter of gene p53 and p16 inarsenic-exposed people with and without malignancy.Toxicol Sci 2006;89:43137.

    99 Chen WT, Hung WC, Kang WY, Huang YC, Chai CY.Urothelial carcinomas arising in arsenic-contaminatedareas are associated with hypermethylation of the genepromoter of the death-associated protein kinase.Histopathology 2007;51:78592.

    100 Marsit CJ, Karagas MR, Danaee H et al. Carcinogen ex-posure and gene promoter hypermethylation in bladdercancer. Carcinogenesis 2006;27:11216.

    101 Zhang AH, Bin HH, Pan XL, Xi XG. Analysis of p16 genemutation, deletion and methylation in patients witharseniasis produced by indoor unventilated-stove coalusage in Guizhou, China. J Toxicol Environ Health A2007;70:97075.

    102 Boonchai W, Walsh M, Cummings M, Chenevix-Trench G. Expression of p53 in arsenic-related and spor-

    adic basal cell carcinoma. Arch Dermatol 2000;136:19598.

    103 Arrigo AP. Acetylation and methylation patterns of corehistones are modified after heat or arsenite treatment ofDrosophila tissue culture cells. Nucleic Acids Res 1983;11:1389404.

    104 Jo WJ, Ren X, Chu F et al. Acetylated H4K16 by MYST1protects UROtsa cells from arsenic toxicity and isdecreased following chronic arsenic exposure. ToxicolAppl Pharmacol 2009;241:294302.

    105 Li J, Chen P, Sinogeeva N et al. Arsenic trioxide promoteshistone H3 phosphoacetylation at the chromatin ofCASPASE-10 in acute promyelocytic leukemia cells.J Biol Chem 2002;277:4950410.

    106 Li J, Gorospe M, Barnes J, Liu Y. Tumor pro-

    moter arsenite stimulates histone H3 phosphoacetyla-tion of proto-oncogenes c-fos and c-jun chromatin inhuman diploid fibroblasts. J Biol Chem 2003;278:1318391.

    107 Ramirez T, Brocher J, Stopper H, Hock R. Sodiumarsenite modulates histone acetylation, histone deacety-lase activity and HMGN protein dynamics in humancells. Chromosoma 2008;117:14757.

    108 Desrosiers R, Tanguay RM. Further characterization ofthe posttranslational modifications of core histones inresponse to heat and arsenite stress in Drosophila.Biochem Cell Biol 1986;64:75057.

    96 INTERNATIONAL JOURNAL OF EPIDEMIOLOGY

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    19/27

    109 Desrosiers R, Tanguay RM. Methylation of Drosophilahistones at proline, lysine, and arginine residues duringheat shock. J Biol Chem 1988;263:468692.

    110 Zhou X, Sun H, Ellen TP, Chen H, Costa M. Arsenitealters global histone H3 methylation. Carcinogenesis2008;29:183136.

    111 Zhou X, Li Q, Arita A, Sun H, Costa M. Effects of nickel,chromate, and arsenite on histone 3 lysine methylation.Toxicol Appl Pharmacol 2009;236:7884.

    112 Zykova TA, Zhu F, Lu C et al. Lymphokine-activatedkiller T-cell-originated protein kinase phosphorylationof histone H2AX prevents arsenite-induced apoptosis inRPMI7951 melanoma cells. Clin Cancer Res 2006;12:688493.

    113 Marsit CJ, Eddy K, Kelsey KT. MicroRNA responses tocellular stress. Cancer Res 2006;66:1084348.

    114 Binet F, Antoine F, Girard D. Interaction between ar-senic trioxide and human primary cells: emphasis onhuman cells of myeloid origin. Inflamm Allergy DrugTargets 2009;8:2127.

    115 Cao Y, Yu SL, Wang Y, Guo GY, Ding Q, An RH.MicroRNA-dependent regulation of PTEN after arsenictrioxide treatment in bladder cancer cell line T24.

    Tumour Biol 2011;32:17988.116 Lee YW, Klein CB, Kargacin B et al. Carcinogenic nickel

    silences gene-expression by chromatin condensation andDNA methylation - a new model for epigenetic carcino-gens. Mol Cell Biol 1995;15:254757.

    117 Lee YW, Klein CB, Kargacin B et al. Carcinogenic nickelsilences gene expression by chromatin condensation andDNA methylation: a new model for epigenetic carcino-gens. Mol Cell Biol 1995;15:254757.

    118 Klein CB, Conway K, Wang XW et al. Senescence ofnickel-transformed cells by an X chromosome: possibleepigenetic control. Science 1991;251:79699.

    119 Govindarajan B, Klafter R, Miller MS et al. Reactiveoxygen-induced carcinogenesis causes hypermethylationof p16(Ink4a) and activation of MAP kinase. Mol Med

    2002;8:18.120 Karaczyn AA, Golebiowski F, Kasprzak KS. Truncation,

    deamidation, and oxidation of histone H2B in cellscultured with nickel(II). Chem Res Toxicol 2005;18:193442.

    121 Karaczyn A, Ivanov S, Reynolds M, Zhitkovich A,Kasprzak KS, Salnikow K. Ascorbate depletion mediatesup-regulation of hypoxia-associated proteins by celldensity and nickel. J Cell Biochem 2006;97:102535.

    122 Broday L, Peng W, Kuo MH, Salnikov K, Zoroddu M,Costa M. Nickel compounds are novel inhibitors of his-tone H4 acetylation. Cancer Res 2000;60:23841.

    123 Chen H, Ke Q, Kluz T, Yan Y, Costa M. Nickel ions in-crease histone H3 lysine 9 dimethylation and inducetransgene silencing. Mol Cell Biol 2006;26:372837.

    124 Ke QD, Davidson T, Chen HB, Kluz T, Costa M.Alterations of histone modifications and transgene silen-cing by nickel chloride. Carcinogenesis 2006;27:148188.

    125 Golebiowski F, Kasprzak KS. Inhibition of core histonesacetylation by carcinogenic nickel(II). Mol Cell Biochem2005;279:13339.

    126 Klein CB, Costa M. DNA methylation, heterochromatinand epigenetic carcinogens. Mutat Res-Rev Mutat Res 1997;386:16380.

    127 Klein CB, Conway K, Wang XW et al. Senescence ofnickel-transformed cells by an x-chromosome - possibleepigenetic control. Science 1991;251:79699.

    128 Yan Y, Kluz T, Zhang P, Chen HB, Costa M. Analysis ofspecific lysine histone H3 and H4 acetylation and methy-lation status in clones of cells with a gene silenced bynickel exposure. Toxicol Appl Pharmacol 2003;190:27277.

    129 Zhang QW, Salnikow K, Kluz T, Chen LC, Su WC,Costa M. Inhibition and reversal of nickel-induced trans-formation by the histone deacetylase inhibitor trichosta-tin A. Toxicol Appl Pharmacol 2003;192:20111.

    130 Broday L, Peng W, Kuo MH, Salnikow K, Zoroddu M,Costa M. Nickel compounds are novel inhibitors of his-tone H4 acetylation. Cancer Res 2000;60:23841.

    131 Golebiowski F, Kasprzak KS. Inhibition of core histonesacetylation by carcinogenic nickel(II). Mol Cell Biochem2005;279:13339.

    132 Ke Q, Li Q, Ellen TP, Sun H, Costa M. Nickel compoundsinduce phosphorylation of histone H3 at serine 10 byactivating JNK-MAPK pathway. Carcinogenesis 2008;29:127681.

    133 Huang D, Zhang Y, Qi Y, Chen C, Ji W. Global DNAhypomethylation, rather than reactive oxygen species(ROS), a potential facilitator of cadmium-stimulatedK562 cell proliferation. Toxicol Lett 2008;179:4347.

    134 Takiguchi M, Achanzar WE, Qu W, Li G, Waalkes MP.

    Effects of cadmium on DNA-(Cytosine-5) methyltrans-ferase activity and DNA methylation status duringcadmium-induced cellular transformation. Exp Cell Res2003;286:35565.

    135 Tellez-Plaza M, Navas-Acien A, Crainiceanu CM,Guallar E. Cadmium exposure and hypertension in the19992004 National Health and Nutrition ExaminationSurvey (NHANES). Environ Health Perspect 2008;116:5156.

    136 Bhatnagar A. Environmental cardiology - Studyingmechanistic links between pollution and heart disease.Circulation Res 2006;99:692705.

    137 Bollati V, Marinelli B, Apostoli P et al. Exposure tometal-rich particulate matter modifies the expression ofcandidate microRNAs in peripheral blood leukocytes.

    Environ Health Perspect 2010;118:76368.138 Williams AE, Perry MM, Moschos SA, Larner-

    Svensson HM, Lindsay MA. Role of miRNA-146a inthe regulation of the innate immune response andcancer. Biochem Soc Trans 2008;36(Pt 6):121115.

    139 Pilsner JR, Lazarus AL, Nam DH et al.Mercury-associated DNA hypomethylation in polar bearbrains via the LUminometric Methylation Assay: a sen-sitive method to study epigenetics in wildlife. Mol Ecol2010;19:30714.

    140 Arai Y, Ohgane J, Yagi S et al. Epigenetic Assessment ofenvironmental chemicals detected in maternal peripheraland cord blood samples. J Reprod Dev 2011;57:50717.

    141 Wright RO, Schwartz J, Wright RJ et al. Biomarkers oflead exposure and DNA methylation within retrotrans-

    posons. Environ Health Perspect 2010;118:79095.142 Pilsner JR, Hu H, Ettinger A et al. Influence of prenatal

    lead exposure on genomic methylation of cord bloodDNA. Environ Health Perspect 2009;117:146671.

    143 Kondo K, Takahashi Y, Hirose Y et al. The reduced ex-pression and aberrant methylation of p16(INK4a) inchromate workers with lung cancer. Lung Cancer 2006;53:295302.

    144 Takahashi Y, Kondo K, Hirose T et al. Microsatellite in-stability and protein expression of the DNA mismatchrepair gene, hMLH1, of lung cancer in chromate-exposedworkers. Mol Carcinogenesis 2005;42:15058.

    ENVIRONMENTAL EPIGENETICS 97

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    20/27

    145 Frouin H, Fortier M, Fournier M. Toxic effects of variouspollutants in 11B7501 lymphoma B cell line from har-bour seal (Phoca vitulina). Toxicology 2010;270:6676.

    146 Apostoli P, Catalani S. Mechanisms of action for metallicelements and their species classified carcinogen R 45 andR 49 by EU. G Ital Med Lav Ergon 2008;30:38291.

    147 Schnekenburger M, Talaska G, Puga A. Chromiumcross-links histone deacetylase 1-DNA methyltransferase1 complexes to chromatin, inhibiting histone-remodelingmarks critical for transcriptional activation. Mol Cell Biol2007;27:7089101.

    148 Sun H, Zhou X, Chen H, Li Q, Costa M. Modulation ofhistone methylation and MLH1 gene silencing by hexa-valent chromium. Toxicol Appl Pharmacol 2009;237:25866.

    149 Pogue AI, Li YY, Cui JG et al. Characterization of anNF-kappaB-regulated, miRNA-146a-mediated down-regulation of complement factor H (CFH) in metal-sulfate-stressed human brain cells. J Inorg Biochem2009;103:159195.

    150 Lukiw WJ, Pogue AI. Induction of specific micro RNA(miRNA) species by ROS-generating metal sulfates inprimary human brain cells. J Inorg Biochem 2007;101:

    126569.151 Jones PA, Baylin SB. The epigenomics of cancer. Cell

    2007;128:68392.152 Kim MS, Lee J, Sidransky D. DNA methylation markers

    in colorectal cancer. Cancer Metastasis Rev 2010;29:181206.

    153 Markowitz SD, Bertagnolli MM. Molecular origins ofcancer: Molecular basis of colorectal cancer. N Engl JMed 2009;361:244960.

    154 Anway MD, Cupp AS, Uzumcu M, Skinner MK.Epigenetic transgenerational actions of endocrine dis-ruptors and male fertility. Science 2005;308:146669.

    155 Guerrero-Bosagna C, Settles M, Lucker B, Skinner MK.Epigenetic transgenerational actions of vinclozolin onpromoter regions of the sperm epigenome. PLoS One

    2010;5:e13100.156 Anway MD, Skinner MK. Epigenetic transgenerational

    actions of endocrine disruptors. Endocrinology 2006;147(6 Suppl):S4349.

    157 Zama AM, Uzumcu M. Fetal and neonatal exposure tothe endocrine disruptor methoxychlor causes epigeneticalterations in adult ovarian genes. Endocrinology 2009;150:468191.

    158 Tao L, Yang S, Xie M, Kramer PM, Pereira MA.Hypomethylation and overexpression of c-jun andc-myc protooncogenes and increased DNA methyltrans-ferase activity in dichloroacetic and trichloroacetic acid-promoted mouse liver tumors. Cancer Lett 2000;158:18593.

    159 Tao L, Yang S, Xie M, Kramer PM, Pereira MA. Effect of

    trichloroethylene and its metabolites, dichloroacetic acidand trichloroacetic acid, on the methylation and expres-sion of c-Jun and c-Myc protooncogenes in mouse liver:prevention by methionine. Toxicol Sci 2000;54:399407.

    160 Hathaway G, Proctor N, Hughes J, Fischman M. ProctorAnd Hughes Chemical Hazards of the Workplace. New York:Van Nostrand Reinhold, 1991.

    161 Rusiecki JA, Baccarelli A, Bollati V, Tarantini L,Moore LE, Bonefeld-Jorgensen EC. Global DNA hypo-methylation is associated with high serum-persistent or-ganic pollutants in Greenlandic Inuit. Environ HealthPerspect 2008;116:154752.

    162 Kim KY, Kim DS, Lee SK et al. Association of low-doseexposure to persistent organic pollutants with globalDNA hypomethylation in healthy koreans. EnvironHealth Perspect 2010;118:37074.

    163 Alavanja MC, Bonner MR. Pesticides and human can-cers. Cancer Invest 2005;23:70011.

    164 Weichenthal S, Moase C, Chan P. A review of pesticideexposure and cancer incidence in the AgriculturalHealth Study cohort. Environ Health Perspect 2010;118:111725.

    165 Alavanja MC, Ward MH, Reynolds P. Carcinogenicity ofagricultural pesticides in adults and children. JAgromedicine 2007;12:3956.

    166 Bassil KL, Vakil C, Sanborn M, Cole DC, Kaur JS,Kerr KJ. Cancer health effects of pesticides: systematicreview. Can Fam Physician 2007;53:170411.

    167 Koutros S, Alavanja MC, Lubin JH et al. An update ofcancer incidence in the Agricultural Health Study. JOccup Environ Med 2010;52:1098105.

    168 Waggoner JK, Kullman GJ, Henneberger PK et al.Mortality in the agricultural health study, 19932007.Am J Epidemiol 2011;173:7183.

    169 Song C, Kanthasamy A, Anantharam V, Sun F,

    Kanthasamy AG. Environmental neurotoxic pesticide in-creases histone acetylation to promote apoptosis in dopa-minergic neuronal cells: relevance to epigeneticmechanisms of neurodegeneration. Mol Pharmacol 2010;77:62132.

    170 Song C, Kanthasamy A, Anantharam V, Sun F,Kanthasamy AG. Environmental neurotoxic pesticide in-creases histone acetylation to promote apoptosis in dopa-minergic neuronal cells: relevance to epigeneticmechanisms of neurodegeneration. Mol Pharmacol 2010;77:62132.

    171 Baccarelli A, Cassano PA, Litonjua A et al. Cardiacautonomic dysfunction - effects from particulate airpollution and protection by dietary methyl nutrientsand metabolic polymorphisms. Circulation 2008;117:

    180209.172 Jardim MJ, Fry RC, Jaspers I, Dailey L, Diaz-Sanchez D.

    Disruption of microRNA expression in human airwaycells by diesel exhaust particles is linked totumorigenesis-associated pathways. Environ HealthPerspect 2009;117:174551.

    173 Tarantini L, Bonzini M, Apostoli P et al. Effects of par-ticulate matter on genomic DNA methylation contentand iNOS promoter methylation. Environ Health Perspect2009;117:21722.

    174 Yauk C, Polyzos A, Rowan-Carroll A et al. Germ-line mu-tations, DNA damage, and global hypermethylation inmice exposed to particulate air pollution in an urban/industrial location. Proc Natl Acad Sci USA 2008;105:60510.

    175 Liu J, Ballaney M, Al-alem U et al. Combined inhaleddiesel exhaust particles and allergen exposure altermethylation of T helper genes and IgE productionin vivo. Toxicol Sci 2008;102:7681.

    176 Perera F, Tang WY, Herbstman J et al. Relation of DNAmethylation of 50-CpG island of ACSL3 to transplacentalexposure to airborne polycyclic aromatic hydrocarbonsand childhood asthma. PLoS One 2009;4:e4488.

    177 Cantone L, Nordio F, Hou L et al. Effects of InhalableMetal-rich Air Particles on Histone H3K4 Dimethylationand H3K9 Acetylation in a Cross-sectional Study of SteelWorkers. Environ Health Perspect 2011;119:9649.

    98 INTERNATIONAL JOURNAL OF EPIDEMIOLOGY

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    21/27

    178 Bollati V, Marinelli B, Apostoli P et al. Exposure tometal-rich particulate matter modifies the expression ofcandidate micrornas in peripheral blood leukocytes.Environ Health Perspect 2010.

    179 Smith MT. Advances in understanding benzene healtheffects and susceptibility. Annu Rev Public Health 2010;31:13348 2 p following 148.

    180 Rinsky RA, Hornung RW, Silver SR, Tseng CY. Benzeneexposure and hematopoietic mortality: a long-term epi-demiologic risk assessment. Am J Ind Med 2002;42:47480.

    181 Goldstein BD. Benzene toxicity. Occup Med 1988;3:54154.

    182 Whitworth KW, Symanski E, Coker AL. Childhood lym-phohematopoietic cancer incidence and hazardous airpollutants in southeast Texas, 19952004. EnvironHealth Perspect 2008;116:157680.

    183 Overall evaluations of carcinogenicity: an updating ofIARC Monographs volumes 1 to 42. IARC Monogr EvalCarcinog Risks Hum Suppl 1987;7:1440.

    184 Richardson DB. Temporal variation in the associationbetween benzene and leukemia mortality. EnvironHealth Perspect 2008;116:37074.

    185 CDC. Facts About Benzene. 2005. Available from: http://www.bt.cdc.gov/agent/benzene/basics/facts.asp.

    186 Bollati V, Baccarelli A, Hou L et al. Changes in DNAmethylation patterns in subjects exposed to low-dosebenzene. Cancer Res 2007;67:87680.

    187 Ji Z, Zhang L, Peng V, Ren X, McHale CM, Smith MT. Acomparison of the cytogenetic alterations and globalDNA hypomethylation induced by the benzene metabol-ite, hydroquinone, with those induced by melphalan andetoposide. Leukemia 2010;24:98691.

    188 Gao A, Zuo X, Liu Q, Lu X, Guo W, Tian L. Methylationof PARP-1 promoter involved in the regulation ofbenzene-induced decrease of PARP-1 mRNA expression.Toxicol Lett 2010;195:11418.

    189 Keri RA, Ho SM, Hunt PA, Knudsen KE, Soto AM,

    Prins GS. An evaluation of evidence for the carcino-genic activity of bisphenol A. Reprod Toxicol 2007;24:24052.

    190 Ho SM, Tang WY, de Frausto JB, Prins GS.Developmental exposure to estradiol and bisphenol Aincreases susceptibility to prostate carcinogenesis andepigenetically regulates phosphodiesterase type 4 variant4. Cancer Res 2006;66:562432.

    191 vom Saal FS, Hughes C. An extensive new literatureconcerning low-dose effects of bisphenol A shows theneed for a new risk assessment. Environ Health Perspect2005;113:92633.

    192 Dolinoy DC, Huang D, Jirtle RL. Maternal nutrient sup-plementation counteracts bisphenol A-induced DNAhypomethylation in early development. Proc Natl Acad

    Sci USA 2007;104:1305661.193 Waterland RA. Is epigenetics an important link between

    early life events and adult disease? Horm Res 2009;71(Suppl 1):1316.

    194 Bromer JG, Zhou Y, Taylor MB, Doherty L, Taylor HS.Bisphenol-A exposure in utero leads to epigenetic alter-ations in the developmental programming of uterine es-trogen response. FASEB J 2010;24:227380.

    195 Weng YI, Hsu PY, Liyanarachchi S et al. Epigenetic in-fluences of low-dose bisphenol A in primary humanbreast epithelial cells. Toxicol Appl Pharmacol 2010;248:11121.

    196 Avissar-Whiting M, Veiga KR, Uhl KM et al. Bisphenol Aexposure leads to specific microRNA alterations in pla-cental cells. Reprod Toxicol 2010;29:40106.

    197 Beedanagari SR, Taylor RT, Bui P, Wang F,Nickerson DW, Hankinson O. Role of epigenetic mech-anisms in differential regulation of the dioxin-induciblehuman CYP1A1 and CYP1B1 genes. Mol Pharmacol 2010;78:60816.

    198 Wang J, Zhao YY, Liu H et al. The role of insulin-likegrowth factor-2 gene differentially methylated regions inTCDD-induced malformation. Zhonghua Yi Xue Yi ChuanXue Za Zhi 2007;24:16266.

    199 McClure EA, North CM, Kaminski NE, Goodman JI.Changes in DNA methylation and gene expressionduring 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced suppression of the lipopolysaccharide (LPS)-stimulated IgM response in splenocytes. Toxicol Sci2011;120:33948.

    200 Elyakim E, Sitbon E, Faerman A et al. hsa-miR-191 is acandidate oncogene target for hepatocellular carcinomatherapy. Cancer Res 2010;70:807787.

    201 Beller HR, Tiemeier K. Use of liquid chromatography/tandem mass spectrometry to detect distinctive indica-

    tors of in situ RDX transformation in contaminatedgroundwater. Environ Sci Techno 2002;36:206066.

    202 Zhang B, Pan X. RDX induces aberrant expression ofmicroRNAs in mouse brain and liver. Environ HealthPerspect 2009;117:23140.

    203 Laitman CJ. DES exposure and the aging woman:mothers and daughters. Curr Womens Health Rep 2002;2:39093.

    204 Palmer JR, Hatch EE, Rosenberg CL et al. Risk of breastcancer in women exposed to diethylstilbestrol in utero:prelimiinary results (United States). Cancer Causes Control2002;13:75358.

    205 Hsu PY, Deatherage DE, Rodriguez BAet al. Xenoestrogen-induced epigenetic repression of microRNA-9-3 in breastepithelial cells. Cancer Res 2009;69:593645.

    206 Mages F, Macovschi O, Prigent AF, Fonlupt P. Increasedmethylation of chloroform extractable products and CTP:cholinephosphate cytidylyltransferase in brain mem-brane preparations from triethyltin-intoxicated rats.Pharmacol Toxicol 1989;65:30205.

    207 Pereira MA, Kramer PM, Conran PB, Tao L. Effect ofchloroform on dichloroacetic acid and trichloroaceticacid-induced hypomethylation and expression of thec-myc gene and on their promotion of liver and kidneytumors in mice. Carcinogenesis 2001;22:151119.

    208 Coffin JC, Ge R, Yang S, Kramer PM, Tao L, Pereira MA.Effect of trihalomethanes on cell proliferation and DNAmethylation in female B6C3F1 mouse liver. Toxicol Sci2000;58:24352.

    209 Chlorinated drinking-water. IARC Monogr Eval Carcinog

    Risks Hum 1991;52:45141.210 Uden PC, Miller JW. Chlorinated acids and chloral in

    drinking water. J Am Water Works Assoc 1983;75:52427.211 Coleman WE, Munch JW, Kaylor WH, Streicher RP,

    Ringhand HP, Meier JR. Gas chromatography/mass spec-troscopy analysis of mutagenic extracts of aqueouschlorinated humic acid. A comparison of the by-productsto drinking water contaminants Environ. Sci. Technol.1984;18:67478.

    212 Shi L, Wu J. Epigenetic regulation in mammalian pre-implantation embryo development. Reprod Biol Endocrinol2009;7:5969.

    ENVIRONMENTAL EPIGENETICS 99

    http://www.bt.cdc.gov/agent/benzene/basics/facts.asphttp://www.bt.cdc.gov/agent/benzene/basics/facts.asphttp://www.bt.cdc.gov/agent/benzene/basics/facts.asphttp://www.bt.cdc.gov/agent/benzene/basics/facts.asp
  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    22/27

    213 Morgan HD, Sutherland HG, Martin DI, Whitelaw E.Epigenetic inheritance at the agouti locus in themouse. Nat Genet 1999;23:31418.

    214 Heijmans BT, Tobi EW, Stein AD et al. Persistent epigen-etic differences associated with prenatal exposure tofamine in humans. Proc Natl Acad Sci USA 2008;105:1704649.

    215 Tobi EW, Lumey LH, Talens RP et al. DNA methylationdifferences after exposure to prenatal famine arecommon and timing- and sex-specific. Hum Mol Genet2009;18:404653.

    216 Heijmans BT, Tobi EW, Lumey LH, Slagboom PE. Theepigenome: archive of the prenatal environment.Epigenetics 2009;4:52631.

    217 Terry MB, Ferris JS, Pilsner R et al. Genomic DNAmethylation among women in a multiethnic New YorkCity birth cohort. Cancer Epidemiol Biomarkers Prev 2008;17:230610.

    218 Flom J, Ferris J, Gonzalez K, Santella R, Terry M.Prenatal tobacco smoke exposure and genomewidemethylation in adulthood. Cancer Epidemiol BiomarkersPrev 2011;20:72021.

    219 Guerrero-Preston R, Goldman LR, Brebi-Mieville P et al.

    Global DNA hypomethylation is associated with in uteroexposure to cotinine and perfluorinated alkyl com-pounds. Epigenetics 2010;5:53946.

    220 Maccani MA, Avissar-Whiting M, Banister CE,McGonnigal B, Padbury JF, Marsit CJ. Maternal cigar-ette smoking during pregnancy is associated with down-regulation of miR-16, miR-21, and miR-146a in theplacenta. Epigenetics 2010;5:58389.

    221 Minard ME, Jain AK, Barton MC. Analysis of epigeneticalterations to chromatin during development. Genesis2009;47:55972.

    222 Relton CL, Davey Smith G. Epigenetic epidemiology ofcommon complex disease: prospects for prediction, pre-vention, and treatment. PLoS Med 2010;7:e1000356.

    223 Geneletti S, Gallo V, Porta M, Khoury MJ, Vineis P.

    Assessing causal relationships in genomics: FromBradford-Hill criteria to complex gene-environmentinteractions and directed acyclic graphs. Emerg ThemesEpidemiol 2011;8:522.

    224 Fuks F. DNA methylation and histone modifications:teaming up to silence genes. Curr Opin Genet Dev 2005;15:49095.

    225 Chuang JC, Jones PA. Epigenetics and microRNAs.Pediatr Res 2007;61(5 Pt 2):24R29R.

    226 Sciandrello G, Caradonna F, Mauro M, Barbata G.Arsenic-induced DNA hypomethylation affects chromo-somal instability in mammalian cells. Carcinogenesis 2004;25:41317.

    227 Smith IM, Mydlarz WK, Mithani SK, Califano JA. DNAglobal hypomethylation in squamous cell head and neck

    cancer associated with smoking, alcohol consumptionand stage. Int J Cancer 2007;121:172428.

    228 Roman-Gomez J, Jimenez-Velasco A, Agirre X et al.Promoter hypermethylation and global hypomethylationare independent epigenetic events in lymphoid leukemo-genesis with opposing effects on clinical outcome.Leukemia 2006;20:144548.

    229 Deng G, Nguyen A, Tanaka H et al. Regional hyper-methylation and global hypomethylation are associatedwith altered chromatin conformation and histone acetyl-ation in colorectal cancer. Int J Cancer 2006;118:29993005.

    230 Brothman AR, Swanson G, Maxwell TM et al. Globalhypomethylation is common in prostate cancer cells: aquantitative predictor for clinical outcome? Cancer GenetCytogenet 2005;156:3136.

    231 Shimabukuro M, Sasaki T, Imamura A et al. Globalhypomethylation of peripheral leukocyte DNA in malepatients with schizophrenia: a potential link betweenepigenetics and schizophrenia. J Psychiatr Res 2007;41:104246.

    232 Bagnyukova TV, Luzhna LI, Pogribny IP, Lushchak VI.Oxidative stress and antioxidant defenses in goldfishliver in response to short-term exposure to arsenite.Environ Mol Mutagen 2007;48:65865.

    233 Pilsner JR, Liu X, Ahsan H et al. Folate deficiency, hyper-homocysteinemia, low urinary creatinine, and hypo-methylation of leukocyte DNA are risk factors forarsenic-induced skin lesions. Environ Health Perspect2009;117:25460.

    234 Cheetham S, Tang MJ, Mesak F, Kennecke H, Owen D,Tai IT. SPARC promoter hypermethylation in colorectalcancers can be reversed by 5-Aza-20 deoxycytidine to in-crease SPARC expression and improve therapy response.British J Cancer 2008;98:181019.

    235 Alemayehu A, Sebova K, Fridrichova I. Redundant DNAmethylation in colorectal cancers of Lynch-syndrome pa-tients. Genes Chrom Cancer 2008;47:90614.

    236 Norrie MWA, Hawkins NJ, Todd AV, Meagher AP,OConnor TW, Ward RL. The role of hMLH1 methylationin the development of synchronous sporadic colorectalcarcinomas. Dis Colon Rectum 2002;45:67480.

    237 Minardi D, Lucarini G, Filosa A et al. Prognostic role ofglobal DNA-methylation and histone acetylation in pT1aclear cell renal carcinoma in partial nephrectomy speci-mens. J Cell Mol Med 2009;13:211521.

    238 Schafer E, Irizarry R, Negi S et al. Promoter hypermethy-lation in MLL-r infant acute lymphoblastic leukemia:biology and therapeutic targeting. Blood 2010;115:4798809.

    239 Owen HC, Giedl J, Wild PJ et al. Low frequency of epi-genetic events in urothelial tumors in young patients.J Urol 2010;184:45963.

    240 Qian J, Yao DM, Lin J et al. Alteration of methy-lation status of death-associated protein kinase(dapk) gene promoter in patients with acute myeloidleukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2010;18:139094.

    241 Laytragoon-Lewin N, Chen F, Castro J et al. DNA contentand methylation of p16, DAPK and RASSF1A gene intumour and distant, normal mucosal tissue of headand neck squamous cell carcinoma patients. AnticancerRes 2010;30:464348.

    242 Paluszczak J, Misiak P, Wierzbicka M, Wozniak A, Baer-Dubowska W. Frequent hypermethylation of DAPK,

    RARbeta, MGMT, RASSF1A and FHIT in laryngeal squa-mous cell carcinomas and adjacent normal mucosa. OralOncol 2011;47:1047.

    243 Hafner N, Diebolder H, Jansen L, Hoppe I, Durst M,Runnebaum IB. Hypermethylated DAPK in serum DNAof women with uterine leiomyoma is a biomarker notrestricted to cancer. Gynecol Oncol 2011;121:2249.

    244 Li B, Wang B, Niu LJ, Jiang L, Qiu CC. Hypermethylationof multiple tumor-related genes associated withDMNT3b up-regulation served as a biomarker for earlydiagnosis of esophageal squamous cell carcinoma.Epigenetics 2011;6:30716.

    100 INTERNATIONAL JOURNAL OF EPIDEMIOLOGY

  • 7/27/2019 Enviromental Chemical Exposure and Human Epigenetics

    23/27

    245 Ben Ayed-Guerfali D, Benhaj K, Khabir A et al.Hypermethylation of tumor-related genes in tunisian pa-tients with gastric carcinoma: Clinical and biological sig-nificance. J Surg Oncol 2011;103:68794.

    246 Sugita H, Iida S, Inokuchi M et al. Methylation of BNIP3and DAPK indicates lower response to chemotherapyand poor prognosis in gastric cancer. Oncol Rep 2011;25:51318.

    247 Zhang Y, Wang R, Song H et al. Methylation of multiplegenes as a candidate biomarker in non-small cell lungcancer. Cancer Lett 2011;303:218.

    248 Hu SL, Kong XY, Cheng ZD