1932

Abstract

DNA methylation is the most well studied of the epigenetic regulators in relation to environmental exposures. To date, numerous studies have detailed the manner by which DNA methylation is influenced by the environment, resulting in altered global and gene-specific DNA methylation. These studies have focused on prenatal, early-life, and adult exposure scenarios. The present review summarizes currently available literature that demonstrates a relationship between DNA methylation and environmental exposures. It includes studies on aflatoxin B, air pollution, arsenic, bisphenol A, cadmium, chromium, lead, mercury, polycyclic aromatic hydrocarbons, persistent organic pollutants, tobacco smoke, and nutritional factors. It also addresses gaps in the literature and future directions for research. These gaps include studies of mixtures, sexual dimorphisms with respect to environmentally associated methylation changes, tissue specificity, and temporal stability of the methylation marks.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-publhealth-040617-014629
2018-04-01
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/publhealth/39/1/annurev-publhealth-040617-014629.html?itemId=/content/journals/10.1146/annurev-publhealth-040617-014629&mimeType=html&fmt=ahah

Literature Cited

  1. Agodi A, Barchitta M, Quattrocchi A, Maugeri A, Canto C. 1.  et al. 2015. Low fruit consumption and folate deficiency are associated with LINE-1 hypomethylation in women of a cancer-free population. Genes Nutr 10:30 [Google Scholar]
  2. Alegría-Torres JA, Carrizales-Yánez L, Díaz-Barriga F, Rosso-Camacho F, Motta V. 2.  et al. 2016. DNA methylation changes in Mexican children exposed to arsenic from two historic mining areas in San Luis Potosí. Environ. Mol. Mutagen. 57:717–23 [Google Scholar]
  3. Ali AH, Kondo K, Namura T, Senba Y, Takizawa H. 3.  et al. 2011. Aberrant DNA methylation of some tumor suppressor genes in lung cancers from workers with chromate exposure. Mol. Carcinog. 50:89–99 [Google Scholar]
  4. Alvarado-Cruz I, Sánchez-Guerra M, Hernández-Cadena L, De Vizcaya-Ruiz A, Mugica V. 4.  et al. 2017. Increased methylation of repetitive elements and DNA repair genes is associated with higher DNA oxidation in children in an urbanized, industrial environment. Mutat. Res. 813:27–36 [Google Scholar]
  5. Amarasekera M, Martino D, Ashley S, Harb H, Kesper D. 5.  et al. 2014. Genome-wide DNA methylation profiling identifies a folate-sensitive region of differential methylation upstream of ZFP57-imprinting regulator in humans. FASEB J 28:4068–76 [Google Scholar]
  6. Ameer SS, Engström K, Hossain MB, Concha G, Vahter M, Broberg K. 6.  2017. Arsenic exposure from drinking water is associated with decreased gene expression and increased DNA methylation in peripheral blood. Toxicol. Appl. Pharmacol. 321:57–66 [Google Scholar]
  7. Appleton AA, Armstrong DA, Lesseur C, Lee J, Padbury JF. 7.  et al. 2013. Patterning in placental 11-B hydroxysteroid dehydrogenase methylation according to prenatal socioeconomic adversity. PLOS ONE 8:e74691 [Google Scholar]
  8. Argos M. 8.  2015. Arsenic exposure and epigenetic alterations: recent findings based on the Illumina 450K DNA methylation array. Curr. Environ. Health Rep. 2:137–44 [Google Scholar]
  9. Argos M, Chen L, Jasmine F, Tong L, Pierce BL. 9.  et al. 2015. Gene-specific differential DNA methylation and chronic arsenic exposure in an epigenome-wide association study of adults in Bangladesh. Environ. Health Perspect. 123:64–71 [Google Scholar]
  10. Armstrong DA, Lesseur C, Conradt E, Lester BM, Marsit CJ. 10.  2014. Global and gene-specific DNA methylation across multiple tissues in early infancy: implications for children's health research. FASEB J 28:2088–97 [Google Scholar]
  11. Azzi S, Sas TC, Koudou Y, Le Bouc Y, Souberbielle J-C. 11.  et al. 2014. Degree of methylation of ZAC1 (PLAGL1) is associated with prenatal and post-natal growth in healthy infants of the EDEN mother child cohort. Epigenetics 9:338–45 [Google Scholar]
  12. Baccarelli A, Wright RO, Bollati V, Tarantini L, Litonjua AA. 12.  et al. 2009. Rapid DNA methylation changes after exposure to traffic particles. Am. J. Respir. Crit. Care Med. 179:572–78 [Google Scholar]
  13. Bailey KA, Wu MC, Ward WO, Smeester L, Rager JE. 13.  et al. 2013. Arsenic and the epigenome: interindividual differences in arsenic metabolism related to distinct patterns of DNA methylation. J. Biochem. Mol. Toxicol. 27:106–15 [Google Scholar]
  14. Bakulski KM, Lee H, Feinberg JI, Wells EM, Brown S. 14.  et al. 2015. Prenatal mercury concentration is associated with changes in DNA methylation at TCEANC2 in newborns. Int. J. Epidemiol. 44:1249–62 [Google Scholar]
  15. Bandyopadhyay AK, Paul S, Adak S, Giri AK. 15.  2016. Reduced LINE-1 methylation is associated with arsenic-induced genotoxic stress in children. Biometals 29:731–41 [Google Scholar]
  16. Beach SRH, Lei MK, Ong ML, Brody GH, Dogan MV, Philibert RA. 16.  2017. MTHFR methylation moderates the impact of smoking on DNA methylation at AHRR for African American young adults. Am. J. Med. Genet. B Neuropsychiatr. Genet. 174:608–18 [Google Scholar]
  17. Bednarska-Makaruk M, Graban A, Sobczyńska-Malefora A, Harrington DJ, Mitchell M. 17.  et al. 2016. Homocysteine metabolism and the associations of global DNA methylation with selected gene polymorphisms and nutritional factors in patients with dementia. Exp. Gerontol. 81:83–91 [Google Scholar]
  18. Bellavia A, Urch B, Speck M, Brook RD, Scott JA. 18.  et al. 2013. DNA hypomethylation, ambient particulate matter, and increased blood pressure: findings from controlled human exposure experiments. J. Am. Heart Assoc. 2:e000212 [Google Scholar]
  19. Bind MA, Baccarelli A, Zanobetti A, Tarantini L, Suh H. 19.  et al. 2012. Air pollution and markers of coagulation, inflammation, and endothelial function: associations and epigene-environment interactions in an elderly cohort. Epidemiology 23:332–40 [Google Scholar]
  20. Bind MA, Lepeule J, Zanobetti A, Gasparrini A, Baccarelli A. 20.  et al. 2014. Air pollution and gene-specific methylation in the Normative Aging Study: association, effect modification, and mediation analysis. Epigenetics 9:448–58 [Google Scholar]
  21. Binder AM, Michels KB. 21.  2013. The causal effect of red blood cell folate on genome-wide methylation in cord blood: a Mendelian randomization approach. BMC Bioinformat. 14:353 [Google Scholar]
  22. Boeke CE, Baccarelli A, Kleinman KP, Burris HH, Litonjua AA. 22.  et al. 2012. Gestational intake of methyl donors and global LINE-1 DNA methylation in maternal and cord blood: prospective results from a folate-replete population. Epigenetics 7:253–60 [Google Scholar]
  23. Bollati V, Favero C, Albetti B, Tarantini L, Moroni A. 23.  et al. 2014. Nutrients intake is associated with DNA methylation of candidate inflammatory genes in a population of obese subjects. Nutrients 6:4625–39 [Google Scholar]
  24. Borghol N, Suderman M, McArdle W, Racine A, Hallett M. 24.  et al. 2012. Associations with early-life socio-economic position in adult DNA methylation. Int. J. Epidemiol. 41:62–74 [Google Scholar]
  25. Braithwaite EC, Kundakovic M, Ramchandani PG, Murphy SE, Champagne FA. 25.  2015. Maternal prenatal depressive symptoms predict infant NR3C1 1F and BDNF IV DNA methylation. Epigenetics 10:408–17 [Google Scholar]
  26. Breton CV, Byun HM, Wenten M, Pan F, Yang A, Gilliland FD. 26.  2009. Prenatal tobacco smoke exposure affects global and gene-specific DNA methylation. Am. J. Respir. Crit. Care Med. 180:462–67 [Google Scholar]
  27. Breton CV, Siegmund KD, Joubert BR, Wang X, Qui W. 27.  et al. 2014. Prenatal tobacco smoke exposure is associated with childhood DNA CpG methylation. PLOS ONE 9:e99716 [Google Scholar]
  28. Breton CV, Yao J, Millstein J, Gao L, Siegmund KD. 28.  et al. 2016. Prenatal air pollution exposures, DNA methyl transferase genotypes, and associations with newborn LINE1 and Alu methylation and childhood blood pressure and carotid intima-media thickness in the Children's Health Study. Environ. Health Perspect. 124:1905–12 [Google Scholar]
  29. Broberg K, Ahmed S, Engström K, Hossain MB, Jurkovic Mlakar S. 29.  et al. 2014. Arsenic exposure in early pregnancy alters genome-wide DNA methylation in cord blood, particularly in boys. J. Dev. Origins Health Dis. 5:288–98 [Google Scholar]
  30. Brocato J, Costa M. 30.  2013. Basic mechanics of DNA methylation and the unique landscape of the DNA methylome in metal-induced carcinogenesis. Crit. Rev. Toxicol. 43:493–514 [Google Scholar]
  31. Byun HM, Motta V, Panni T, Bertazzi PA, Apostoli P. 31.  et al. 2013. Evolutionary age of repetitive element subfamilies and sensitivity of DNA methylation to airborne pollutants. Part. Fibre Toxicol. 10:28 [Google Scholar]
  32. Byun HM, Panni T, Motta V, Hou L, Nordio F. 32.  et al. 2013. Effects of airborne pollutants on mitochondrial DNA methylation. Part. Fibre Toxicol. 10:18 [Google Scholar]
  33. Cardenas A, Houseman EA, Baccarelli AA, Quamruzzaman Q, Rahman M. 33.  et al. 2015. In utero arsenic exposure and epigenome-wide associations in placenta, umbilical artery, and human umbilical vein endothelial cells. Epigenetics 10:1054–63 [Google Scholar]
  34. Cardenas A, Koestler DC, Houseman EA, Jackson BP, Kile ML. 34.  et al. 2015. Differential DNA methylation in umbilical cord blood of infants exposed to mercury and arsenic in utero. Epigenetics 10:508–15 [Google Scholar]
  35. Cardenas A, Rifas-Shiman SL, Agha G, Hivert M-F, Litonjua AA. 35.  et al. 2017. Persistent DNA methylation changes associated with prenatal mercury exposure and cognitive performance during childhood. Sci. Rep. 7:288 [Google Scholar]
  36. Carmona JJ, Sofer T, Hutchinson J, Cantone L, Coull B. 36.  et al. 2014. Short-term airborne particulate matter exposure alters the epigenetic landscape of human genes associated with the mitogen-activated protein kinase network: a cross-sectional study. Environ. Health 13:94 [Google Scholar]
  37. Cheng L, Liu J, Li B, Liu S, Li X, Tu H. 37.  2016. Cigarette smoke-induced hypermethylation of the GCLC gene is associated with COPD. Chest 149:474–82 [Google Scholar]
  38. Chung J-Y, Yu S-D, Hong Y-S. 38.  2014. Environmental source of arsenic exposure. J. Prev. Med. Public Health 47:253–57 [Google Scholar]
  39. Clifford RL, Jones MJ, MacIsaac JL, McEwen LM, Goodman SJ. 39.  et al. 2017. Inhalation of diesel exhaust and allergen alters human bronchial epithelium DNA methylation. J. Allergy Clin. Immunol. 139:112–21 [Google Scholar]
  40. Consales C, Toft G, Leter G, Bonde JP, Uccelli R. 40.  et al. 2016. Exposure to persistent organic pollutants and sperm DNA methylation changes in Arctic and European populations. Environ. Mol. Mutagen. 57:200–9 [Google Scholar]
  41. Conway K, Edmiston SN, Parrish E, Bryant C, Tse CK. 41.  et al. 2017. Breast tumor DNA methylation patterns associated with smoking in the Carolina Breast Cancer Study. Breast Cancer Res. Treat. 163:349–61 [Google Scholar]
  42. Corrales J, Kristofco LA, Steele WB, Yates BS, Breed CS. 42.  et al. 2015. Global assessment of bisphenol A in the environment: review and analysis of its occurrence and bioaccumulation. Dose-Response 13:1559325815598308 [Google Scholar]
  43. Dao T, Hong X, Wang X, Tang WY. 43.  2015. Aberrant 5′-CpG methylation of cord blood TNFα associated with maternal exposure to polybrominated diphenyl ethers. PLOS ONE 10:e0138815 [Google Scholar]
  44. De Prins S, Koppen G, Jacobs G, Dons E, Van de Mieroop E. 44.  et al. 2013. Influence of ambient air pollution on global DNA methylation in healthy adults: a seasonal follow-up. Environ. Int. 59:418–24 [Google Scholar]
  45. Ding R, Jin Y, Liu X, Ye H, Zhu Z. 45.  et al. 2017. Dose- and time- effect responses of DNA methylation and histone H3K9 acetylation changes induced by traffic-related air pollution. Sci. Rep. 7:43737 [Google Scholar]
  46. Dolinoy DC. 46.  2008. The agouti mouse model: an epigenetic biosensor for nutritional and environmental alterations on the fetal epigenome. Nutr. Rev. 66:Suppl. 1S7–11 [Google Scholar]
  47. Drobná Z, Martin E, Kim KS, Smeester L, Bommarito P. 47.  et al. 2016. Analysis of maternal polymorphisms in arsenic (+3 oxidation state)-methyltransferase AS3MT and fetal sex in relation to arsenic metabolism and infant birth outcomes: implications for risk analysis. Reprod. Toxicol. 61:28–38 [Google Scholar]
  48. Duan H, He Z, Ma J, Zhang B, Sheng Z. 48.  et al. 2013. Global and MGMT promoter hypomethylation independently associated with genomic instability of lymphocytes in subjects exposed to high-dose polycyclic aromatic hydrocarbon. Arch. Toxicol. 87:2013–22 [Google Scholar]
  49. Dunaway KW, Islam MS, Coulson RL, Lopez SJ, Vogel Ciernia A. 49.  et al. 2016. Cumulative impact of polychlorinated biphenyl and large chromosomal duplications on DNA methylation, chromatin, and expression of autism candidate genes. Cell Rep 17:3035–48 [Google Scholar]
  50. Engström K, Wojdacz TK, Marabita F, Ewels P, Käller M. 50.  et al. 2017. Transcriptomics and methylomics of CD4-positive T cells in arsenic-exposed women. Arch. Toxicol. 91:2067–78 [Google Scholar]
  51. Everson TM, Armstrong DA, Jackson BP, Green BB, Karagas MR, Marsit CJ. 51.  2016. Maternal cadmium, placental PCDHAC1, and fetal development. Reprod. Toxicol. 65:263–71 [Google Scholar]
  52. Faulk C, Kim JH, Anderson OS, Nahar MS, Jones TR. 52.  et al. 2016. Detection of differential DNA methylation in repetitive DNA of mice and humans perinatally exposed to bisphenol A. Epigenetics 11:489–500 [Google Scholar]
  53. Faulk C, Kim JH, Jones TR, McEachin RC, Nahar MS. 53.  et al. 2015. Bisphenol A-associated alterations in genome-wide DNA methylation and gene expression patterns reveal sequence-dependent and non-monotonic effects in human fetal liver. Environ. Epigenet. 1:dvv006 [Google Scholar]
  54. Ferrario D, Gribaldo L, Hartung T. 54.  2016. Arsenic exposure and immunotoxicity: a review including the possible influence of age and sex. Curr. Environ. Health Rep. 3:1–12 [Google Scholar]
  55. Flom JD, Ferris JS, Liao Y, Tehranifar P, Richards CB. 55.  et al. 2011. Prenatal smoke exposure and genomic DNA methylation in a multiethnic birth cohort. Cancer Epidemiol. Biomark. Prev. 20:2518–23 [Google Scholar]
  56. Fraga MF, Esteller M. 56.  2007. Epigenetics and aging: the targets and the marks. Trends Genet 23:413–18 [Google Scholar]
  57. Fryer AA, Emes RD, Ismail KM, Haworth KE, Mein C. 57.  et al. 2011. Quantitative, high-resolution epigenetic profiling of CpG loci identifies associations with cord blood plasma homocysteine and birth weight in humans. Epigenetics 6:86–94 [Google Scholar]
  58. Fryer AA, Nafee TM, Ismail KM, Carroll WD, Emes RD, Farrell WE. 58.  2009. LINE-1 DNA methylation is inversely correlated with cord plasma homocysteine in man: a preliminary study. Epigenetics 4:394–98 [Google Scholar]
  59. Gilliland FD, Harms HJ, Crowell RE, Li Y-F, Willink R, Belinsky SA. 59.  2002. Glutathione S-transferase P1 and NADPH quinone oxidoreductase polymorphisms are associated with aberrant promoter methylation of P16INK4a and O6-methylguanine-DNA methyltransferase in sputum. Cancer Res 62:2248–52 [Google Scholar]
  60. Godfrey KM, Sheppard A, Gluckman PD, Lillycrop KA, Burdge GC. 60.  et al. 2011. Epigenetic gene promoter methylation at birth is associated with child's later adiposity. Diabetes 60:1528–34 [Google Scholar]
  61. Gonseth S, Roy R, Houseman EA, de Smith AJ, Zhou M. 61.  et al. 2015. Periconceptional folate consumption is associated with neonatal DNA methylation modifications in neural crest regulatory and cancer development genes. Epigenetics 10:1166–76 [Google Scholar]
  62. Goodrich JM, Basu N, Franzblau A, Dolinoy DC. 62.  2013. Mercury biomarkers and DNA methylation among Michigan dental professionals. Environ. Mol. Mutagen. 54:195–203 [Google Scholar]
  63. Goodrich JM, Reddy P, Naidoo RN, Asharam K, Batterman S, Dolinoy DC. 63.  2016. Prenatal exposures and DNA methylation in newborns: a pilot study in Durban, South Africa. Environ. Sci. Process. Impacts 18:908–17 [Google Scholar]
  64. Goodrich JM, Sánchez BN, Dolinoy DC, Zhang Z, Hernandez-Ávila M. 64.  et al. 2015. Quality control and statistical modeling for environmental epigenetics: a study on in utero lead exposure and DNA methylation at birth. Epigenetics 10:19–30 [Google Scholar]
  65. Green BB, Karagas MR, Punshon T, Jackson BP, Robbins DJ. 65.  et al. 2016. Epigenome-wide assessment of DNA methylation in the placenta and arsenic exposure in the New Hampshire Birth Cohort Study (USA). Environ. Health Perspect. 124:1253–60 [Google Scholar]
  66. Gudsnuk K, Champagne FA. 66.  2012. Epigenetic influence of stress and the social environment. ILAR J 53:279–88 [Google Scholar]
  67. Guerrero-Preston R, Goldman LR, Brebi-Mieville P, Ili-Gangas C, Lebron C. 67.  et al. 2010. Global DNA hypomethylation is associated with in utero exposure to cotinine and perfluorinated alkyl compounds. Epigenetics 5:539–46 [Google Scholar]
  68. Gunter TD, Philibert RA. 68.  2015. Smoking, methylation at AHRR, and recidivism risk in a community correction sample of individuals at high risk for recidivism. Behav. Sci. Law 33:691–700 [Google Scholar]
  69. Guo L, Byun H-M, Zhong J, Motta V, Barupal J. 69.  et al. 2014. Effects of short-term exposure to inhalable particulate matter on DNA methylation of tandem repeats. Environ. Mol. Mutagen. 55:322–35 [Google Scholar]
  70. Hanna CW, Bloom MS, Robinson WP, Kim D, Parsons PJ. 70.  et al. 2012. DNA methylation changes in whole blood is associated with exposure to the environmental contaminants, mercury, lead, cadmium and bisphenol A, in women undergoing ovarian stimulation for IVF. Hum. Reprod. 27:1401–10 [Google Scholar]
  71. Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ. 71.  et al. 2008. Persistent epigenetic differences associated with prenatal exposure to famine in humans. PNAS 105:17046–49 [Google Scholar]
  72. Herbstman JB, Tang D, Zhu D, Qu L, Sjodin A. 72.  et al. 2012. Prenatal exposure to polycyclic aromatic hydrocarbons, benzo[α]pyrene-DNA adducts, and genomic DNA methylation in cord blood. Environ. Health Perspect. 120:733–38 [Google Scholar]
  73. Hernandez-Vargas H, Castelino J, Silver MJ, Dominguez-Salas P, Cros MP. 73.  et al. 2015. Exposure to aflatoxin B1 in utero is associated with DNA methylation in white blood cells of infants in The Gambia. Int. J. Epidemiol. 44:1238–48 [Google Scholar]
  74. Hew KM, Walker AI, Kohli A, Garcia M, Syed A. 74.  et al. 2015. Childhood exposure to ambient polycyclic aromatic hydrocarbons is linked to epigenetic modifications and impaired systemic immunity in T cells. Clin. Exp. Allergy 45:238–48 [Google Scholar]
  75. Ho SM, Johnson A, Tarapore P, Janakiram V, Zhang X, Leung YK. 75.  2012. Environmental epigenetics and its implication on disease risk and health outcomes. ILAR J 53:289–305 [Google Scholar]
  76. Hossain K, Suzuki T, Hasibuzzaman MM, Islam MS, Rahman A. 76.  et al. 2017. Chronic exposure to arsenic, LINE-1 hypomethylation, and blood pressure: a cross-sectional study in Bangladesh. Environ. Health 16:20 [Google Scholar]
  77. Hossain MB, Vahter M, Concha G, Broberg K. 77.  2012. Environmental arsenic exposure and DNA methylation of the tumor suppressor gene p16 and the DNA repair gene MLH1: effect of arsenic metabolism and genotype. Metallomics 4:1167–75 [Google Scholar]
  78. Hossain MB, Vahter M, Concha G, Broberg K. 78.  2012. Low-level environmental cadmium exposure is associated with DNA hypomethylation in Argentinean women. Environ. Health Perspect. 120:879–84 [Google Scholar]
  79. Hou L, Zhang X, Zheng Y, Wang S, Dou C. 79.  et al. 2014. Altered methylation in tandem repeat element and elemental component levels in inhalable air particles. Environ. Mol. Mutagen. 55:256–65 [Google Scholar]
  80. Hoyo C, Daltveit AK, Iversen E, Benjamin-Neelon SE, Fuemmeler B. 80.  et al. 2014. Erythrocyte folate concentrations, CpG methylation at genomically imprinted domains, and birth weight in a multiethnic newborn cohort. Epigenetics 9:1120–30 [Google Scholar]
  81. Hoyo C, Murtha AP, Schildkraut JM, Jirtle RL, Demark-Wahnefried W. 81.  et al. 2011. Methylation variation at IGF2 differentially methylated regions and maternal folic acid use before and during pregnancy. Epigenetics 6:928–36 [Google Scholar]
  82. Huen K, Yousefi P, Bradman A, Yan L, Harley KG. 82.  et al. 2014. Effects of age, sex, and persistent organic pollutants on DNA methylation in children. Environ. Mol. Mutagen. 55:209–22 [Google Scholar]
  83. Inbar-Feigenberg M, Choufani S, Butcher DT, Roifman M, Weksberg R. 83.  2013. Basic concepts of epigenetics. Fertil. Steril. 99:607–15 [Google Scholar]
  84. Intarasunanont P, Navasumrit P, Waraprasit S, Chaisatra K, Suk WA. 84.  et al. 2012. Effects of arsenic exposure on DNA methylation in cord blood samples from newborn babies and in a human lymphoblast cell line. Environ. Health 11:31 [Google Scholar]
  85. Itoh H, Iwasaki M, Kasuga Y, Yokoyama S, Onuma H. 85.  et al. 2014. Association between serum organochlorines and global methylation level of leukocyte DNA among Japanese women: a cross-sectional study. Sci. Total Environ. 490:603–9 [Google Scholar]
  86. Ivorra C, Fraga MF, Bayón GF, Fernández AF, Garcia-Vicent C. 86.  et al. 2015. DNA methylation patterns in newborns exposed to tobacco in utero. J. Transl. Med. 13:25 [Google Scholar]
  87. Janssen BG, Byun HM, Gyselaers W, Lefebvre W, Baccarelli AA, Nawrot TS. 87.  2015. Placental mitochondrial methylation and exposure to airborne particulate matter in the early life environment: An ENVIRONAGE birth cohort study. Epigenetics 10:536–44 [Google Scholar]
  88. Janssen BG, Godderis L, Pieters N, Poels K, Kiciński M. 88.  et al. 2013. Placental DNA hypomethylation in association with particulate air pollution in early life. Part Fibre Toxicol. 10:22 [Google Scholar]
  89. Janssen BG, Gyselaers W, Byun H-M, Roels HA, Cuypers A. 89.  et al. 2017. Placental mitochondrial DNA and CYP1A1 gene methylation as molecular signatures for tobacco smoke exposure in pregnant women and the relevance for birth weight. J. Transl. Med. 15:5 [Google Scholar]
  90. Jimenez-Garza O, Baccarelli AA, Byun HM, Marquez-Gamino S, Barron-Vivanco BS, Albores A. 90.  2015. CYP2E1 epigenetic regulation in chronic, low-level toluene exposure: relationship with oxidative stress and smoking habit. Toxicol. Appl. Pharmacol. 286:207–15 [Google Scholar]
  91. Jin S, Lee YK, Lim YC, Zheng Z, Lin XM. 91.  et al. 2013. Global DNA hypermethylation in down syndrome placenta. PLOS Genet. 9:e1003515 [Google Scholar]
  92. Jones PA. 92.  2012. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 13:484–92 [Google Scholar]
  93. Joubert BR, den Dekker HT, Felix JF, Bohlin J, Ligthart S. 93.  et al. 2016. Maternal plasma folate impacts differential DNA methylation in an epigenome-wide meta-analysis of newborns. Nat. Commun. 7:10577 [Google Scholar]
  94. Joubert BR, Håberg SE, Nilsen RM, Wang X, Vollset SE. 94.  et al. 2012. 450K epigenome-wide scan identifies differential DNA methylation in newborns related to maternal smoking during pregnancy. Environ. Health Perspect. 120:1425–31 [Google Scholar]
  95. Jung KH, Torrone D, Lovinsky-Desir S, Perzanowski M, Bautista J. 95.  et al. 2017. Short-term exposure to PM2.5 and vanadium and changes in asthma gene DNA methylation and lung function decrements among urban children. Respir. Res. 18:63 [Google Scholar]
  96. Kappil MA, Li Q, Li A, Dassanayake PS, Xia Y. 96.  et al. 2016. In utero exposures to environmental organic pollutants disrupt epigenetic marks linked to fetoplacental development. Environ. Epigenet. 2:dvv013 [Google Scholar]
  97. Kile ML, Baccarelli A, Hoffman E, Tarantini L, Quamruzzaman Q. 97.  et al. 2012. Prenatal arsenic exposure and DNA methylation in maternal and umbilical cord blood leukocytes. Environ. Health Perspect. 120:1061–66 [Google Scholar]
  98. Kile ML, Fang S, Baccarelli AA, Tarantini L, Cavallari J, Christiani DC. 98.  2013. A panel study of occupational exposure to fine particulate matter and changes in DNA methylation over a single workday and years worked in boilermaker welders. Environ. Health 12:47 [Google Scholar]
  99. Kim JH, Rozek LS, Soliman AS, Sartor MA, Hablas A. 99.  et al. 2013. Bisphenol A-associated epigenomic changes in prepubescent girls: a cross-sectional study in Gharbiah, Egypt. Environ. Health 12:33 [Google Scholar]
  100. Kim K-Y, Kim D-S, Lee S-K, Lee I-K, Kang J-H. 100.  et al. 2010. Association of low-dose exposure to persistent organic pollutants with global DNA hypomethylation in healthy Koreans. Environ. Health Perspect. 118:370–74 [Google Scholar]
  101. Kim YH, Lee YS, Lee DH, Kim DS. 101.  2016. Polycyclic aromatic hydrocarbons are associated with insulin receptor substrate 2 methylation in adipose tissues of Korean women. Environ. Res. 150:47–51 [Google Scholar]
  102. King K, Murphy S, Hoyo C. 102.  2015. Epigenetic regulation of newborns' imprinted genes related to gestational growth: patterning by parental race/ethnicity and maternal socioeconomic status. J. Epidemiol. Community Health 69:639–47 [Google Scholar]
  103. Kippler M, Engström K, Mlakar SJ, Bottai M, Ahmed S. 103.  et al. 2013. Sex-specific effects of early life cadmium exposure on DNA methylation and implications for birth weight. Epigenetics 8:494–503 [Google Scholar]
  104. Kobayashi S, Azumi K, Goudarzi H, Araki A, Miyashita C. 104.  et al. 2017. Effects of prenatal perfluoroalkyl acid exposure on cord blood IGF2/H19 methylation and ponderal index: the Hokkaido Study. J. Expo. Sci. Environ. Epidemiol. 27:251–59 [Google Scholar]
  105. Koestler DC, Avissar-Whiting M, Houseman EA, Karagas MR, Marsit CJ. 105.  2013. Differential DNA methylation in umbilical cord blood of infants exposed to low levels of arsenic in utero. Environ. Health Perspect. 121:971–77 [Google Scholar]
  106. Kohli RM, Zhang Y. 106.  2013. TET enzymes, TDG and the dynamics of DNA demethylation. Nature 502:472–79 [Google Scholar]
  107. Kok DE, Dhonukshe-Rutten RA, Lute C, Heil SG, Uitterlinden AG. 107.  et al. 2015. The effects of long-term daily folic acid and vitamin B12 supplementation on genome-wide DNA methylation in elderly subjects. Clin. Epigenetics 7:121 [Google Scholar]
  108. Ladd-Acosta C, Shu C, Lee BK, Gidaya N, Singer A. 108.  et al. 2016. Presence of an epigenetic signature of prenatal cigarette smoke exposure in childhood. Environ. Res. 144:139–48 [Google Scholar]
  109. Lam LL, Emberly E, Fraser HB, Neumann SM, Chen E. 109.  et al. 2012. Factors underlying variable DNA methylation in a human community cohort. PNAS 109:Suppl. 217253–60 [Google Scholar]
  110. Lambrou A, Baccarelli A, Wright RO, Weisskopf M, Bollati V. 110.  et al. 2012. Arsenic exposure and DNA methylation among elderly men. Epidemiology 23:668–76 [Google Scholar]
  111. Lee DH, Jacobs DR Jr., Porta M. 111.  2009. Hypothesis: a unifying mechanism for nutrition and chemicals as lifelong modulators of DNA hypomethylation. Environ. Health Perspect. 117:1799–802 [Google Scholar]
  112. Lee J, Kalia V, Perera F, Herbstman J, Li T. 112.  et al. 2017. Prenatal airborne polycyclic aromatic hydrocarbon exposure, LINE1 methylation and child development in a Chinese cohort. Environ. Int. 99:315–20 [Google Scholar]
  113. Leter G, Consales C, Eleuteri P, Uccelli R, Specht IO. 113.  et al. 2014. Exposure to perfluoroalkyl substances and sperm DNA global methylation in Arctic and European populations. Environ. Mol. Mutagen. 55:591–600 [Google Scholar]
  114. Li C, Yang X, Xu M, Zhang J, Sun N. 114.  2013. Epigenetic marker (LINE-1 promoter) methylation level was associated with occupational lead exposure. Clin. Toxicol. 51:225–29 [Google Scholar]
  115. Li W, Deng J, Wang SS, Ma L, Pei J. 115.  et al. 2014. Association of methylation of the RAR-β gene with cigarette smoking in non-small cell lung cancer with Southern-Central Chinese population. Asian Pac. J. Cancer Prevent. 15:10937–41 [Google Scholar]
  116. Lind L, Penell J, Luttropp K, Nordfors L, Syvänen AC. 116.  et al. 2013. Global DNA hypermethylation is associated with high serum levels of persistent organic pollutants in an elderly population. Environ. Int. 59:456–61 [Google Scholar]
  117. Loucks EB, Huang YT, Agha G, Chu S, Eaton CB. 117.  et al. 2016. Epigenetic mediators between childhood socioeconomic disadvantage and mid-life body mass index: the New England Family Study. Psychosom. Med. 78:1053–65 [Google Scholar]
  118. Maccani JZ, Koestler DC, Houseman EA, Marsit CJ, Kelsey KT. 118.  2013. Placental DNA methylation alterations associated with maternal tobacco smoking at the RUNX3 gene are also associated with gestational age. Epigenomics 5:619–30 [Google Scholar]
  119. Maccani JZ, Koestler DC, Lester B, Houseman EA, Armstrong DA. 119.  et al. 2015. Placental DNA methylation related to both infant toenail mercury and adverse neurobehavioral outcomes. Environ. Health Perspect. 123:723–29 [Google Scholar]
  120. Madrigano J, Baccarelli A, Mittleman MA, Wright RO, Sparrow D. 120.  et al. 2011. Prolonged exposure to particulate pollution, genes associated with glutathione pathways, and DNA methylation in a cohort of older men. Environ. Health Perspect. 119:977–82 [Google Scholar]
  121. Majumdar S, Chanda S, Ganguli B, Mazumder DN, Lahiri S, Dasgupta UB. 121.  2010. Arsenic exposure induces genomic hypermethylation. Environ. Toxicol. 25:315–18 [Google Scholar]
  122. Mansell T, Novakovic B, Meyer B, Rzehak P, Vuillermin P. 122.  et al. 2016. The effects of maternal anxiety during pregnancy on IGF2/H19 methylation in cord blood. Transl. Psychiatry 6:e765 [Google Scholar]
  123. Markunas CA, Xu Z, Harlid S, Wade PA, Lie RT. 123.  et al. 2014. Identification of DNA methylation changes in newborns related to maternal smoking during pregnancy. Environ. Health Perspect. 122:1147–53 [Google Scholar]
  124. Martin E, Smeester L, Bommarito PA, Grace MR, Boggess K. 124.  et al. 2017. Sexual epigenetic dimorphism in the human placenta: implications for susceptibility during the prenatal period. Epigenomics 9:267–78 [Google Scholar]
  125. Martin EM, Fry RC. 125.  2016. A cross-study analysis of prenatal exposures to environmental contaminants and the epigenome: support for stress-responsive transcription factor occupancy as a mediator of gene-specific CpG methylation patterning. Environ. Epigenet. 2:dvv011 [Google Scholar]
  126. Mathers JC. 126.  2007. Early nutrition: impact on epigenetics. Forum Nutr 60:42–48 [Google Scholar]
  127. McKay JA, Groom A, Potter C, Coneyworth LJ, Ford D. 127.  et al. 2012. Genetic and non-genetic influences during pregnancy on infant global and site specific DNA methylation: role for folate gene variants and vitamin B12. PLOS ONE 7:e33290 [Google Scholar]
  128. Mitchell MM, Woods R, Chi LH, Schmidt RJ, Pessah IN. 128.  et al. 2012. Levels of select PCB and PBDE congeners in human postmortem brain reveal possible environmental involvement in 15q11-q13 duplication autism spectrum disorder. Environ. Mol. Mutagen. 53:589–98 [Google Scholar]
  129. Mohanty AF, Farin FM, Bammler TK, MacDonald JW, Afsharinejad Z. 129.  et al. 2015. Infant sex-specific placental cadmium and DNA methylation associations. Environ. Res. 138:74–81 [Google Scholar]
  130. Montrose L, Ward TJ, Semmens EO, Cho YH, Brown B, Noonan CW. 130.  2017. Dietary intake is associated with respiratory health outcomes and DNA methylation in children with asthma. Allergy Asthma Clin. Immunol. 13:12 [Google Scholar]
  131. Mozhui K, Smith AK, Tylavsky FA. 131.  2015. Ancestry dependent DNA methylation and influence of maternal nutrition. PLOS ONE 10:e0118466 [Google Scholar]
  132. Murphy SK, Huang Z, Hoyo C. 132.  2012. Differentially methylated regions of imprinted genes in prenatal, perinatal and postnatal human tissues. PLOS ONE 7:e40924 [Google Scholar]
  133. Nahar MS, Kim JH, Sartor MA, Dolinoy DC. 133.  2014. Bisphenol A-associated alterations in the expression and epigenetic regulation of genes encoding xenobiotic metabolizing enzymes in human fetal liver. Environ. Mol. Mutagen. 55:184–95 [Google Scholar]
  134. Nahar MS, Liao C, Kannan K, Harris C, Dolinoy DC. 134.  2015. In utero bisphenol A concentration, metabolism, and global DNA methylation across matched placenta, kidney, and liver in the human fetus. Chemosphere 124:54–60 [Google Scholar]
  135. Needham BL, Smith JA, Zhao W, Wang X, Mukherjee B. 135.  et al. 2015. Life course socioeconomic status and DNA methylation in genes related to stress reactivity and inflammation: the multi-ethnic study of atherosclerosis. Epigenetics 10:958–69 [Google Scholar]
  136. Niedzwiecki MM, Hall MN, Liu X, Oka J, Harper KN. 136.  et al. 2013. A dose-response study of arsenic exposure and global methylation of peripheral blood mononuclear cell DNA in Bangladeshi adults. Environ. Health Perspect. 121:1306–12 [Google Scholar]
  137. Niedzwiecki MM, Liu X, Hall MN, Thomas T, Slavkovich V. 137.  et al. 2015. Sex-specific associations of arsenic exposure with global DNA methylation and hydroxymethylation in leukocytes: results from two studies in Bangladesh. Cancer Epidemiol. Biomark. Prev. 24:1748–57 [Google Scholar]
  138. Nye MD, King KE, Darrah TH, Maguire R, Jima DD. 138.  et al. 2016. Maternal blood lead concentrations, DNA methylation of MEG3 DMR regulating the DLK1/MEG3 imprinted domain and early growth in a multiethnic cohort. Environ. Epigenet. 2:dvv009 [Google Scholar]
  139. Olden K, Lin YS, Gruber D, Sonawane B. 139.  2014. Epigenome: biosensor of cumulative exposure to chemical and nonchemical stressors related to environmental justice. Am. J. Public Health 104:1816–21 [Google Scholar]
  140. Ouyang B, Baxter CS, Lam H-M, Yeramaneni S, Levin L. 140.  et al. 2012. Hypomethylation of dual specificity phosphatase 22 promoter correlates with duration of service in firefighters and is inducible by low-dose benzo[a]pyrene. J. Occup. Environ. Med. 54:774–80 [Google Scholar]
  141. Park SY, Kim K-S, Lee Y-M, Kim M-J, Jacobs DR Jr.. 141.  et al. 2015. Persistent organic pollutants and promoter hypermethylation of the O6-methylguanine-DNA methyltransferase gene. Biomarkers 20:136–42 [Google Scholar]
  142. Pauwels S, Duca RC, Devlieger R, Freson K, Straetmans D. 142.  et al. 2016. Maternal methyl-group donor intake and global DNA (hydroxy)methylation before and during pregnancy. Nutrients 8:474 [Google Scholar]
  143. Pauwels S, Ghosh M, Duca RC, Bekaert B, Freson K. 143.  et al. 2017. Dietary and supplemental maternal methyl-group donor intake and cord blood DNA methylation. Epigenetics 12:1–10 [Google Scholar]
  144. Pauwels S, Ghosh M, Duca RC, Bekaert B, Freson K. 144.  et al. 2017. Maternal intake of methyl-group donors affects DNA methylation of metabolic genes in infants. Clin. Epigenet. 9:16 [Google Scholar]
  145. Pauwels S, Truijen I, Ghosh M, Duca RC, Langie SAS. 145.  et al. 2017. The effect of paternal methyl-group donor intake on offspring DNA methylation and birth weight. J. Dev. Origins Health Dis. 8:311–21 [Google Scholar]
  146. Pavanello S, Bollati V, Pesatori AC, Kapka L, Bolognesi C. 146.  et al. 2009. Global and gene-specific promoter methylation changes are related to anti-B[a]PDE-DNA adduct levels and influence micronuclei levels in polycyclic aromatic hydrocarbon-exposed individuals. Int. J. Cancer 125:1692–97 [Google Scholar]
  147. Pavanello S, Pesatori AC, Dioni L, Hoxha M, Bollati V. 147.  et al. 2010. Shorter telomere length in peripheral blood lymphocytes of workers exposed to polycyclic aromatic hydrocarbons. Carcinogenesis 31:216–21 [Google Scholar]
  148. Peluso ME, Munnia A, Bollati V, Srivatanakul P, Jedpiyawongse A. 148.  et al. 2014. Aberrant methylation of hypermethylated-in-cancer-1 and exocyclic DNA adducts in tobacco smokers. Toxicol. Sci. 137:47–54 [Google Scholar]
  149. Perera F, Tang W-Y, Herbstman J, Tang D, Levin L. 149.  et al. 2009. Relation of DNA methylation of 5′-CpG island of ACSL3 to transplacental exposure to airborne polycyclic aromatic hydrocarbons and childhood asthma. PLOS ONE 4:e4488 [Google Scholar]
  150. Philibert RA, Beach SR, Brody GH. 150.  2012. Demethylation of the aryl hydrocarbon receptor repressor as a biomarker for nascent smokers. Epigenetics 7:1331–38 [Google Scholar]
  151. Pilsner JR, Hall MN, Liu X, Ahsan H, Ilievski V. 151.  et al. 2011. Associations of plasma selenium with arsenic and genomic methylation of leukocyte DNA in Bangladesh. Environ. Health Perspect. 119:113–18 [Google Scholar]
  152. Pilsner JR, Hall MN, Liu X, Ilievski V, Slavkovich V. 152.  et al. 2012. Influence of prenatal arsenic exposure and newborn sex on global methylation of cord blood DNA. PLOS ONE 7:e37147 [Google Scholar]
  153. Pilsner JR, Liu X, Ahsan H, Ilievski V, Slavkovich V. 153.  et al. 2007. Genomic methylation of peripheral blood leukocyte DNA: influences of arsenic and folate in Bangladeshi adults. Am. J. Clin. Nutr. 86:1179–86 [Google Scholar]
  154. Pusceddu I, Herrmann M, Kirsch SH, Werner C, Hubner U. 154.  et al. 2016. Prospective study of telomere length and LINE-1 methylation in peripheral blood cells: the role of B vitamins supplementation. Eur. J. Nutr. 55:1863–73 [Google Scholar]
  155. Rager JE, Tilley SK, Tulenko SE, Smeester L, Ray PD. 155.  et al. 2015. Identification of novel gene targets and putative regulators of arsenic-associated DNA methylation in human urothelial cells and bladder cancer. Chem. Res. Toxicol. 28:1144–55 [Google Scholar]
  156. Reichetzeder C, Dwi Putra SE, Pfab T, Slowinski T, Neuber C. 156.  et al. 2016. Increased global placental DNA methylation levels are associated with gestational diabetes. Clin. Epigenet. 8:82 [Google Scholar]
  157. Reynolds LM, Magid HS, Chi GC, Lohman K, Barr RG. 157.  et al. 2017. Secondhand tobacco smoke exposure associations with DNA methylation of the aryl hydrocarbon receptor repressor. Nicotine Tob. Res. 19:442–51 [Google Scholar]
  158. Reynolds LM, Wan M, Ding J, Taylor JR, Lohman K. 158.  et al. 2015. DNA methylation of the aryl hydrocarbon receptor repressor associations with cigarette smoking and subclinical atherosclerosis. Circ. Cardiovasc. Genet. 8:707–16 [Google Scholar]
  159. Rojas D, Rager JE, Smeester L, Bailey KA, Drobná Z. 159.  et al. 2015. Prenatal arsenic exposure and the epigenome: identifying sites of 5-methylcytosine alterations that predict functional changes in gene expression in newborn cord blood and subsequent birth outcomes. Toxicol. Sci. 143:97–106 [Google Scholar]
  160. Rotroff DM, Joubert BR, Marvel SW, Håberg SE, Wu MC. 160.  et al. 2016. Maternal smoking impacts key biological pathways in newborns through epigenetic modification in utero. BMC Genom 17:976 [Google Scholar]
  161. Ruiz-Hernandez A, Kuo CC, Rentero-Garrido P, Tang WY, Redon J. 161.  et al. 2015. Environmental chemicals and DNA methylation in adults: a systematic review of the epidemiologic evidence. Clin. Epigenet. 7:55 [Google Scholar]
  162. Rusiecki JA, Baccarelli A, Bollati V, Tarantini L, Moore LE, Bonefeld-Jorgensen EC. 162.  2008. Global DNA hypomethylation is associated with high serum-persistent organic pollutants in Greenlandic Inuit. Environ. Health Perspect. 116:1547–52 [Google Scholar]
  163. Rusiecki JA, Beane Freeman LE, Bonner MR, Alexander M, Chen L. 163.  et al. 2017. High pesticide exposure events and DNA methylation among pesticide applicators in the agricultural health study. Environ. Mol. Mutagen. 58:19–29 [Google Scholar]
  164. Rzehak P, Saffery R, Reischl E, Covic M, Wahl S. 164.  et al. 2016. Maternal smoking during pregnancy and DNA-methylation in children at age 5.5 years: epigenome-wide-analysis in the European Childhood Obesity Project (CHOP)-study. PLOS ONE 11:e0155554 [Google Scholar]
  165. Saenen ND, Vrijens K, Janssen BG, Roels HA, Neven KY. 165.  et al. 2017. Lower placental leptin promoter methylation in association with fine particulate matter air pollution during pregnancy and placental nitrosative stress at birth in the ENVIRONAGE cohort. Environ. Health Perspect. 125:262–68 [Google Scholar]
  166. Salam MT, Byun HM, Lurmann F, Breton CV, Wang X. 166.  et al. 2012. Genetic and epigenetic variations in inducible nitric oxide synthase promoter, particulate pollution, and exhaled nitric oxide levels in children. J. Allergy Clin. Immunol. 129:232–39.e1–7 [Google Scholar]
  167. Sanchez-Cespedes M, Decker PA, Doffek KM, Esteller M, Westra WH. 167.  et al. 2001. Increased loss of chromosome 9p21 but not p16 inactivation in primary non-small cell lung cancer from smokers. Cancer Res 61:2092–96 [Google Scholar]
  168. Sanchez-Guerra M, Zheng Y, Osorio-Yanez C, Zhong J, Chervona Y. 168.  et al. 2015. Effects of particulate matter exposure on blood 5-hydroxymethylation: results from the Beijing truck driver air pollution study. Epigenetics 10:633–42 [Google Scholar]
  169. Sanders AP, Smeester L, Rojas D, DeBussycher T, Wu MC. 169.  et al. 2014. Cadmium exposure and the epigenome: exposure-associated patterns of DNA methylation in leukocytes from mother-baby pairs. Epigenetics 9:212–21 [Google Scholar]
  170. Schoeters G, Den Hond E, Zuurbier M, Naginiene R, van den Hazel P. 170.  et al. 2006. Cadmium and children: exposure and health effects. Acta Paediatr. Suppl. 95:50–54 [Google Scholar]
  171. Schwender K, Holtkötter H, Johann KS, Glaub A, Schürenkamp M. 171.  et al. 2016. Sudden infant death syndrome: exposure to cigarette smoke leads to hypomethylation upstream of the growth factor independent 1 (GFI1) gene promoter. Forensic Sci. Med. Pathol. 12:399–406 [Google Scholar]
  172. Sen A, Cingolani P, Senut MC, Land S, Mercado-Garcia A. 172.  et al. 2015. Lead exposure induces changes in 5-hydroxymethylcytosine clusters in CpG islands in human embryonic stem cells and umbilical cord blood. Epigenetics 10:607–21 [Google Scholar]
  173. Sen A, Heredia N, Senut MC, Hess M, Land S. 173.  et al. 2015. Early life lead exposure causes gender-specific changes in the DNA methylation profile of DNA extracted from dried blood spots. Epigenomics 7:379–93 [Google Scholar]
  174. Sen A, Heredia N, Senut MC, Land S, Hollocher K. 174.  et al. 2015. Multigenerational epigenetic inheritance in humans: DNA methylation changes associated with maternal exposure to lead can be transmitted to the grandchildren. Sci. Rep. 5:14466 [Google Scholar]
  175. Shenker NS, Ueland PM, Polidoro S, van Veldhoven K, Ricceri F. 175.  et al. 2013. DNA methylation as a long-term biomarker of exposure to tobacco smoke. Epidemiology 24:712–16 [Google Scholar]
  176. Skinner MK. 176.  2015. Environmental epigenetics and a unified theory of the molecular aspects of evolution: a neo-Lamarckian concept that facilitates neo-Darwinian evolution. Genome Biol. Evol. 7:1296–302 [Google Scholar]
  177. Smeester L, Rager JE, Bailey KA, Guan X, Smith N. 177.  et al. 2011. Epigenetic changes in individuals with arsenicosis. Chem. Res. Toxicol. 24:165–67 [Google Scholar]
  178. Somineni HK, Zhang X, Biagini Myers JM, Kovacic MB, Ulm A. 178.  et al. 2016. Ten-eleven translocation 1 (TET1) methylation is associated with childhood asthma and traffic-related air pollution. J. Allergy Clin. Immunol. 137:797–805.e5 [Google Scholar]
  179. Steegers-Theunissen RP, Obermann-Borst SA, Kremer D, Lindemans J, Siebel C. 179.  et al. 2009. Periconceptional maternal folic acid use of 400 microg per day is related to increased methylation of the IGF2 gene in the very young child. PLOS ONE 4:e7845 [Google Scholar]
  180. Subramanyam MA, Diez-Roux AV, Pilsner JR, Villamor E, Donohue KM. 180.  et al. 2013. Social factors and leukocyte DNA methylation of repetitive sequences: the multi-ethnic study of atherosclerosis. PLOS ONE 8:e54018 [Google Scholar]
  181. Suter M, Abramovici A, Showalter L, Hu M, Shope CD. 181.  et al. 2010. In utero tobacco exposure epigenetically modifies placental CYP1A1 expression. Metabolism 59:1481–90 [Google Scholar]
  182. Suter M, Ma J, Harris A, Patterson L, Brown KA. 182.  et al. 2011. Maternal tobacco use modestly alters correlated epigenome-wide placental DNA methylation and gene expression. Epigenetics 6:1284–94 [Google Scholar]
  183. Tao MH, Zhou J, Rialdi AP, Martinez R, Dabek J. 183.  et al. 2014. Indoor air pollution from solid fuels and peripheral blood DNA methylation: findings from a population study in Warsaw, Poland. Environ. Res. 134:325–30 [Google Scholar]
  184. Tellez-Plaza M, Tang WY, Shang Y, Umans JG, Francesconi KA. 184.  et al. 2014. Association of global DNA methylation and global DNA hydroxymethylation with metals and other exposures in human blood DNA samples. Environ. Health Perspect. 122:946–54 [Google Scholar]
  185. Turner PC, Collinson AC, Cheung YB, Gong Y, Hall AJ. 185.  et al. 2007. Aflatoxin exposure in utero causes growth faltering in Gambian infants. Int. J. Epidemiol. 36:1119–25 [Google Scholar]
  186. Ulrich CM, Toriola AT, Koepl LM, Sandifer T, Poole EM. 186.  et al. 2012. Metabolic, hormonal and immunological associations with global DNA methylation among postmenopausal women. Epigenetics 7:1020–28 [Google Scholar]
  187. Vidal AC, Semenova V, Darrah T, Vengosh A, Huang Z. 187.  et al. 2015. Maternal cadmium, iron and zinc levels, DNA methylation and birth weight. BMC Pharmacol. Toxicol. 16:20 [Google Scholar]
  188. Virani S, Rentschler KM, Nishijo M, Ruangyuttikarn W, Swaddiwudhipong W. 188.  et al. 2016. DNA methylation is differentially associated with environmental cadmium exposure based on sex and smoking status. Chemosphere 145:284–90 [Google Scholar]
  189. Wadhwa PD, Buss C, Entringer S, Swanson JM. 189.  2009. Developmental origins of health and disease: brief history of the approach and current focus on epigenetic mechanisms. Semin. Reprod. Med. 27:358–68 [Google Scholar]
  190. Wang C, Chen R, Cai J, Shi J, Yang C. 190.  et al. 2016. Personal exposure to fine particulate matter and blood pressure: a role of angiotensin converting enzyme and its DNA methylation. Environ. Int. 94:661–66 [Google Scholar]
  191. Wang IJ, Chen SL, Lu TP, Chuang EY, Chen PC. 191.  2013. Prenatal smoke exposure, DNA methylation, and childhood atopic dermatitis. Clin. Exp. Allergy 43:535–43 [Google Scholar]
  192. Wang TC, Song YS, Wang H, Zhang J, Yu SF. 192.  et al. 2012. Oxidative DNA damage and global DNA hypomethylation are related to folate deficiency in chromate manufacturing workers. J. Hazard. Mater. 213–214:440–46 [Google Scholar]
  193. Watkins DJ, Wellenius GA, Butler RA, Bartell SM, Fletcher T, Kelsey KT. 193.  2014. Associations between serum perfluoroalkyl acids and LINE-1 DNA methylation. Environ. Int. 63:71–76 [Google Scholar]
  194. White AJ, Chen J, McCullough LE, Xu X, Cho YH. 194.  et al. 2015. Polycyclic aromatic hydrocarbon (PAH)-DNA adducts and breast cancer: modification by gene promoter methylation in a population-based study. Cancer Causes Control 26:1791–802 [Google Scholar]
  195. White AJ, Chen J, Teitelbaum SL, McCullough LE, Xu X. 195.  et al. 2016. Sources of polycyclic aromatic hydrocarbons are associated with gene-specific promoter methylation in women with breast cancer. Environ. Res. 145:93–100 [Google Scholar]
  196. Wilson AS, Power BE, Molloy PL. 196.  2007. DNA hypomethylation and human diseases. Biochim. Biophys. Acta 1775:138–62 [Google Scholar]
  197. Wu HC, Wang Q, Yang HI, Tsai WY, Chen CJ, Santella RM. 197.  2013. Global DNA methylation in a population with aflatoxin B1 exposure. Epigenetics 8:962–69 [Google Scholar]
  198. Yang P, Ma J, Zhang B, Duan H, He Z. 198.  et al. 2012. CpG site-specific hypermethylation of p16INK4α in peripheral blood lymphocytes of PAH-exposed workers. Cancer Epidemiol. Biomark. Prev. 21:182–90 [Google Scholar]
  199. Yang TY, Hsu LI, Chiu AW, Pu YS, Wang SH. 199.  et al. 2014. Comparison of genome-wide DNA methylation in urothelial carcinomas of patients with and without arsenic exposure. Environ. Res. 128:57–63 [Google Scholar]
  200. Zhang Y-J, Wu H-C, Yazici H, Yu M-W, Lee P-H, Santella RM. 200.  2012. Global hypomethylation in hepatocellular carcinoma and its relationship to aflatoxin B1 exposure. World J. Hepatol. 4:169–75 [Google Scholar]
  201. Zhao Y, Liu P, Wang J, Xiao X, Meng X, Zhang Y. 201.  2016. Umbilical cord blood PBDEs concentrations are associated with placental DNA methylation. Environ. Int. 97:1–6 [Google Scholar]
  202. Zhu H, Wang G, Qian J. 202.  2016. Transcription factors as readers and effectors of DNA methylation. Nat. Rev. Genet. 17:551–65 [Google Scholar]
  203. Zhu X, Li J, Deng S, Yu K, Liu X. 203.  et al. 2016. Genome-wide analysis of DNA methylation and cigarette smoking in a Chinese population. Environ. Health Perspect. 124:966–73 [Google Scholar]
/content/journals/10.1146/annurev-publhealth-040617-014629
Loading
/content/journals/10.1146/annurev-publhealth-040617-014629
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error