BibTex RIS Cite

Epigenetik Etkileri Olan Endokrin Bozucu Kimyasal Maddeler: Bisfenol A

Year 2016, Issue: 2, 186 - 203, 01.06.2016

Abstract

Son yıllarda epigenetik mekanizmaların gen ifade değişikliklerine neden olduğu ve bu değişikliklerin de başta kanser olmak üzere birçok hastalığın ortaya çıkmasında rol oynadığı çok sayıda kapsamlı çalışma ile gösterilmiştir. İnsanların yaygın olarak maruz kaldığı bazı çevresel kirleticilerin epigenetik değişikliklere neden olarak bazı hastalıkların patogenezinde rol oynadıklarını gösteren veriler bulunmaktadır. Bisfenol A BPA en yaygın maruz kalınan çevresel kirleticilerden olup, başta plastik sanayi olmak üzere endüstride pek çok alanda kullanımı olan bir kimyasal maddedir. BPA’nın yaşamın farklı dönemlerinde farklı epigenetik etkilerinin bulunduğu ve bu etkilerin fertilite ve kanser gelişimi gibi geniş bir yelpazede sonuçlarının olduğu bilinmektedir. Bu derleme kapsamında epigenetik mekanizmalar incelenecektir ve BPA’nın epigenetik etkilerinin değerlendirildiği in vitro ve in vivo çalışmaların sonuçları sunulacaktır.

References

  • Jiang Y, Bressler J, Beaudet LA: Epigenetics and human disease. Annual Reviews of Genetics 2004, 5:479-510.
  • Chatterjee R, Vinson C: CpG methylation recruits sequence specific transcription factors essential for tissue specific gene expression. Biochimica et Biophysica Acta. 2012, 1819(7):763-770.
  • Deaton AM, Bird A: CpG islands and the regulation of transcription. Genes and Development 2011, 25(10):1010-1022.
  • Kulis M, Queirós AC, Beekman R, Martín-Subero JI: Intragenic DNA methylation in transcriptional regulation, normal differentiation and cancer. Biochimica et Biophysica Acta 2013, 1829(11):1161-1174.
  • Meng H, Cao Y, Qin J, Song X, Zhang Q, Shi Y, Cao L: DNA methylation, its mediators and genome integrity. International Journal of Biological Sciences 2015, 11(5):604-617.
  • Erdmann A, Halby L, Fahy J, Arimondo PB: Targeting DNA methylation with small molecules: what’s next? Journal of Medicinal Chemistry 2015, 58(6):2569-2583.
  • Du J, Johnson LM, Jacobsen SE, Patel DJ: DNA methylation pathways and their crosstalk with histone methylation. Nature Reviews Molecular Cell Biology 2015, 16(9):519-532.
  • Dunn BK. Hypomethylation: one side of a larger picture. Annals of the New York Academy of Sciences. 2003, 983:28-42.
  • Wilson AS, Power BE, Molloy PL: DNA hypomethylation and human diseases. Biochim Biophys Acta. 2007, 1775(1):138-162.
  • Watanabe Y, Maekawa M: Methylation of DNA in cancer. Advances in Clinical Chemistry 2010, 52:145-167.
  • Zelic R, Fiano V, Grasso C, Zugna D, Pettersson A, Gillio-Tos A, Merletti F, Richiardi L: Global DNA hypomethylation in prostate cancer development and progression: a systematic review. Prostate Cancer and Prostatic Diseases 2015, 18(1):1-12.
  • Heichman KA, Warren JD: DNA methylation biomarkers and their utility for solid cancer diagnosticsClinical Chemistry and Laboratory Medicine 2012, 50(10):1707-1721.
  • Gokul G, Khosla S. DNA methylation and cancer. Subcell Biochem. 2013, 61:597-625.
  • Brennan K, Flanagan JM: Is there a link between genome-wide hypomethylation in blood and cancer risk? Cancer Prevention Research (Phila). 2012, 5(12):1345-1357.
  • Baba Y, Watanabe M, Baba H: Review of the alterations in DNA methylation in esophageal squamous cell carcinoma. Surgery Today 2013, 43(12):1355-1364.
  • Akhavan-Niaki H, Samadani AA: DNA methylation and cancer development: molecular mechanism. Cell Biochem Biophys. 2013, 67(2):501-513.
  • Woloszynska-Read A, Zhang W, Yu J, Link PA, Mhawech-Fauceglia P, Collamat G, Akers SN, Ostler KR, Godley LA, Odunsi K, Karpf AR: Coordinated cancer germline antigen promoter and global DNA hypomethylation in ovarian cancer: association with the BORIS/CTCF expression ratio and advanced stage. Clinical Cancer Research 2011, 17(8):2170-2180.
  • Lam K, Pan K, Linnekamp JF, Medema JP, Kandimalla R: DNA methylation based biomarkers in colorectal cancer: A systematic review. Biochimica et Biophysica Acta 2016, 1866(1):106-120.
  • Segre CV, Chiocca S: Regulating the regulators: The Post-Translational Code of Class I HDAC1 and HDAC2. Journal of Biomedicine and Biotechnology 2011, 2011:690848.
  • Moazed D, Bühler M, Buker SM, Colmenares SU, Gerace EL, Gerber SA, Hong EJ, Motamedi MR, Verdel A, Villén J, Gygi SP: Studies on the mechanism of RNAi-dependent heterochromatin assembly. Cold Spring Harbor Symposia on Quantitative Biology 2006, 71:461-471.
  • Agrawal N, Dasaradhi PV, Mohmmed A, Malhotra P, Bhatnagar RK, Mukherjee SK: RNA interference: biology, mechanism, and applications. Microbiology and Molecular Biology Reviews 2003, 67(4):657-685.
  • Egger G, Liang G, Aparicio A, Jones AP: Epigenetics in human disease and prospects for epigenetic therapy. Nature 2004, 429:457-463.
  • Ghildiyal M, Zamore PD: Small silencing RNAs: an expanding universe. Nature Reviews Genetics 2009, 10(2):94-108.
  • Pratt AJ, MacRae IJ: The RNA-induced silencing complex: a versatile gene-silencing machine. Journal of Biological Chemistry 2009, 284(27):17897-17901.
  • Kim YJ, Maizel A, Chen X: Traffic into silence: endomembranes and post-transcriptional RNA silencing. EMBO Journal 2014, 33(9):968-980.
  • Bajrami E, Spiroski M: Genomic Imprinting. Open Access Macedonian Journal of Medical Sciences 2016, 4(1):181-184.
  • Nicholls RD, Knepper JL: Genome organization, function, and imprinting in Prader-Willi and Angelman syndromes. Annu Rev Genomics Human Genetics 2001;2:153-175.
  • Jacob KJ, Robinson WP, Lefebvre L: Beckwith-Wiedemann and Silver-Russell syndromes: opposite developmental imbalances in imprinted regulators of placental function and embryonic growth. Clinşcal Genetics 2013, 84(4):326-334.
  • Marczak-Hałupka A, Kalina MA, Tańska A, Chrzanowska KH: Silver-Russell Syndrome - Part I: Clinical Characteristics and Genetic Background. Pediatric Endocrinology, Diabetes and Metabolism 2015, 20(3):101-106.
  • Levine MA: An update on the clinical and molecular characteristics of pseudohypoparathyroidism. Curr Opinions in Endocrinology, Diabetes and Obesity 2012, 19(6):443-451.
  • Herrera BM, Keildson S, Lindgren CM: Genetics and epigenetics of obesity. Maturitas. 2011, 69(1):41-49.
  • Pfeifer K: Mechanisms of Genomic Imprinting American Journal of Human Genetics, 2000, 67:777-877
  • Sleutels F, Barlow DP, Lyle R: The uniqueness of the imprinting mechanism. Current Opinions in Genetics and Development 2000,10:229-233.
  • Fundele RH, Surani MA, Allen ND. Consequences of genomic imprinting for fetal development. In: Reik W, Surani A (eds), Genomic imprinting, IRL/Oxford University Press, Oxford, New York, Tokyo 1997: 98–112.
  • Lintelmann J, Katayama A, Kurıhara N, Shore L, Wenzel A: Endocrine disrupters in the environment IUPAC Technical Report, Pure and Applied Chemistry 2003, 75(5):631-681.
  • Staples CA, Dorn PB, Klecka GM, O’Block ST, Harris LR: A review of the environmental fate, effects, and exposures of bisphenol A. Chemosphere 1998, 36(10):2149-2173.
  • Vom Saal FS, Nagel SC, Coe BL, Angle BM, Taylor JA. The estrogenic endocrine disrupting chemical bisphenol A (BPA) and obesity. Molecular and Cellular Endocrinology 2012, 354(1-2):74-84.
  • Johnson S, Saxena P, Sahu R: Leaching of Bisphenol A from Baby Bottles. Proceedings of the National Academy of Sciences, India Section B: Biological Sciences 2015, 85(1):131-135.
  • Olea N, Pulgar R, Pérez P, Olea-Serrano F, Rivas A, Novillo-Fertrell A, Pedraza V, Soto AM, Sonnenschein C: Estrogenicity of resin-based composites and sealants in dentistry. Environmental Health Perspectives 1996,104(3):298-305.
  • Vandenberg LN, Hunt PA, Myers JP, Vom Saal FS: Human exposures to bisphenol A: mismatches between data and assumptions. Reviews in Environmental Health 2013, 28(1):37-58.
  • Fenichel P, Chevalier N, Brucker-Davis F: Bisphenol A: an endocrine and metabolic disruptor. Annuals of Endocrinology (Paris) 2013, 74(3):211-220.
  • Batra T: Bisphenol-A in canned food products: is it really required? Archives of Industrial Hygiene and Toxicology 2011, 62(4):381-384.
  • Tyl RW, Myers CB, Marr MC, Thomas BF, Keimowitz AR, Brine DR, Veselica MM, Fail PA, Chang TY, Seely JC, Joiner RL, Butala JH, Dimond SS, Cagen SZ, Shiotsuka RN, Stropp GD, Waechter JM: Three-generation reproductive toxicity study of dietary bisphenol A in CD Sprague- Dawley rats. Toxicological Sciences 2002, 68(1):121-146.
  • Weng YI, Hsu PY, Liyanarachchi S, Liu J, Deatherage DE, Huang YW, Zuo T, Rodriguez B, Lin CH, Cheng AL, Huang TH: Epigenetic influences of low-dose bisphenol A in primary human breast epithelial cells. Toxicology and Applied Pharmacology 2010, 248(2):111-121.
  • Nagelkerke A, Sieuwerts AM, Bussink J, Sweep FC, Look MP, Foekens JA, Martens JW, Span PN: LAMP3 is involved in tamoxifen resistance in breast cancer cells through the modulation of autophagy. Endocrine-Related Cancer. 2014, 21(1):101-112.
  • Doherty LF, Bromer JG, Zhou Y, Aldad TS, Taylor HS: In utero exposure to diethylstilbestrol (DES) or bisphenol-A (BPA) increases EZH2 expression in the mammary gland: an epigenetic mechanism linking endocrine disruptors to breast cancer. Hormones and Cancer 2010, 1(3):146-155.
  • Bhan A, Hussain I, Ansari KI, Bobzean SA, Perrotti LI, Mandal SS. Histone methyltransferase EZH2 is transcriptionally induced by estradiol as well as estrogenic endocrine disruptors bisphenol-A and diethylstilbestrol. Journal of Molecular0 Biology 2014, 426(20):3426-3441.
  • Avissar-Whiting M, Veiga KR, Uhl KM, Maccani MA, Gagne LA, Moen EL, Marsit CJ: Bisphenol A exposure leads to specific microRNA alterations in placental cells. Reproductive Toxicology 2010, 29(4):401-406.
  • Cho H, Kim S, Park H.-W, Oh M-J, Yu S, Lee S, Park C, Han J, Oh J-H, Hwang S, Yoon S-J: A relationship between miRNA and gene expression in the mouse sertoli cell line after exposure to bisphenol A. BioChip Journal 2010, 4(1):75–81.
  • Wang T, Han J, Duan X, Xiong B, Cui XS, Kim NH, Liu HL, Sun SC: The toxic effects and possible mechanisms of Bisphenol A on oocyte maturation of porcine in vitro. Oncotarget 2016, 7(22):32554-32565.
  • Yaoi T, Itoh K, Nakamura K, Ogi H, Fujiwara Y, Fushiki S: Genome-wide analysis of epigenomic alterations in fetal mouse forebrain after exposure to low doses of bisphenol A. Biochemical and Biophysical Research Communications 2008, 376(3):563–567.
  • Dolinoy DC, Huang D, Jirtle RL: Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development. Proceedings of National Academecy of Sciences USA. 2007, 104(32):13056–13061.
  • Kochmanski J, Marchlewicz EH, Savidge M, Montrose L, Faulk C, Dolinoy DC: Longitudinal effects of developmental bisphenol A and variable diet exposures on epigenetic drift in mice. Reproductive Toxicology 2016, pii: S0890-6238(16)30308-2.
  • Manikkam M, Guerrero-Bosagna C, Tracey R, Haque MM, Skinner MK: Transgenerational actions of environmental compounds on reproductive disease and identification of epigenetic biomarkers of ancestral exposures. PLoS One. 2012, 7:e31901.
  • Doshi T, Mehta SS, Dighe V, Balasinor N, Vanage G: Hypermethylation of estrogen receptor promoter region in adult testis of rats exposed neonatally to bisphenol A. Toxicology. 2011;289(2- 3):74–82.
  • Mao Z, Xia W, Chang H, Huo W, Li Y, Xu S: Paternal BPA exposure in early life alters Igf2 epigenetic status in sperm and induces pancreatic impairment in rat offspring. Toxicology Letters 2015 238(3):30-38.
  • Ho SM, Tang WY: Belmonte de Frausto J, Prins GS: Developmental exposure to estradiol and bisphenol a increases susceptibility to prostate carcinogenesis and epigenetically regulates phosphodiesterase type 4 variant 4. Cancer Research 2006, 66(11):5624–5632.
  • Prins GS, Tang WY, Belmonte J, Ho SM: Developmental exposure to bisphenol a increases prostate cancer susceptibility in adult rats: Epigenetic mode of action is implicated. Fertility and Sterility 2008, 89:e41.
  • Cheong A, Zhang X, Cheung YY, Tang WY, Chen J, Ye SH, Medvedovic M, Leung YK, Prins GS, Ho SM: DNA methylome changes by estradiol benzoate and bisphenol A links early-life environmental exposures to prostate cancer risk. Epigenetics 2016. DOI: 10.1080/15592294.2016.1208891.
  • Weinhouse C, Sartor MA, Faulk C, Anderson OS, Sant KE, Harris C, Dolinoy DC: Epigenome- wide DNA methylation analysis implicates neuronal and inflammatory signaling pathways in adult murine hepatic tumorigenesis following perinatal exposure to bisphenol A. Environmental and Molecular Mutagenesis 2016, 57(6):435-446.
  • Deb P, Bhan A, Hussain I, Ansari KI, Bobzean SA, Pandita TK, Perrotti LI, Mandal SS: Endocrine disrupting chemical, bisphenol-A, induces breast cancer associated gene HOXB9 expression in vitro and in vivo. Gene 2016, 590(2):234-243.
  • Lamartiniere CA, Jenkins S, Betancourt AM, Wang J, Russo J: Exposure to the Endocrine Disruptor Bisphenol A Alters Susceptibility for Mammary Cancer. Hormone Molecular Biology and Clinical Investigation 2011, 5(2):45-52.
  • Bhan A, Hussain I, Ansari KI, Bobzean SA, Perrotti LI, Mandal SS: Bisphenol-A and diethylstilbestrol exposure induces the expression of breast cancer associated long noncoding RNA HOTAIR in vitro and in vivo. Journal Of Steroid Biochemistry And Molecular Biology 2014, 141:160-170.
  • Dhimolea E, Wadia PR, Murray TJ, Settles ML, Treitman JD, Sonnenschein C, Shioda T, Soto AM: Prenatal exposure to BPA alters the epigenome of the rat mammary gland and increases the propensity to neoplastic development. PLoS One 2014, 9(7):e99800.
  • Greathouse KL, Bredfeldt T, Everitt JI, Lin K, Berry T, Kannan K, Mittelstadt ML, Ho SM, Walker CL: Environmental estrogens differentially engage the histone methyltransferase EZH2 to increase risk of uterine tumorigenesis. Molecular Cancer Research 2012, 10(4):546-557.
  • Faulk C, Kim JH, Jones TR, McEachin RC, Nahar MS, Dolinoy DC, Sartor MA: Bisphenol A-associated alterations in genome-wide DNA methylation and gene expression patterns reveal sequence-dependent and non-monotonic effects in human fetal liver. Environmental Epigenetics 2015, 1(1). pii: dvv006.
  • Faulk C, Kim JH, Anderson OS, Nahar MS, Jones TR, Sartor MA, Dolinoy DC: Detection of differential DNA methylation in repetitive DNA of mice and humans perinatally exposed to bisphenol A. Epigenetics. 2016, 11(7):489-500.
  • Kundakovic M, Champagne FA: Epigenetic perspective on the developmental effects of bisphenol A. Brain Behavior and Immunity 2011, 25(6):1084–1093.
  • Jones BA, Shimell JJ, Watson NV. Pre- and postnatal bisphenol A treatment results in persistent deficits in the sexual behavior of male rats, but not female rats, in adulthood. Hormones and Behavior 2011, 59(2):246-251.
  • Prins GS, Tang WY, Belmonte J, Ho SM. Perinatal exposure to oestradiol and bisphenol A alters the prostate epigenome and increases susceptibility to carcinogenesis. Basic Clinical Pharmacology and Toxicology 2008, 102(2):134-138.
  • Yin N, Yao X, Qin Z, Wang YL, Faiola F: Assessment of Bisphenol A (BPA) neurotoxicity in vitro with mouse embryonic stem cells. Journal of Environmetal Sciences (China) 2015, 36:181-187.
  • Li J, Fu KZ, Vemula S, Le XC, Li XF. Studying developmental neurotoxic effects of bisphenol A (BPA) using embryonic stem cells. Journal of Environmetal Sciences (China) 2015, 1;36:173-177.
  • Hessel EV, Ezendam J, van Broekhuizen FA, Hakkert B, DeWitt J, Granum B, Guzylack L, Lawrence BP, Penninks A, Rooney AA, Piersma AH, van Loveren H. Assessment of recent developmental immunotoxicity studies with bisphenol A in the context of the 2015 EFSA TDI. Reproductive Toxicology 2016, 65:448-456.
  • Robinson L, Miller R. The Impact of Bisphenol A and Phthalates on Allergy, Asthma, and Immune Function: a Review of Latest Findings. Current Environmental Health Reports 2015, 2(4):379-387.
  • Ziv-Gal A, Flaws JA. Evidence for bisphenol A-induced female infertility: a review (2007-2016). Fertility and Sterility 2016, 106(4):827-856.
  • Seachrist DD, Bonk KW, Ho SM, Prins GS, Soto AM, Keri RA. A review of the carcinogenic potential of bisphenol A. Reproductive Toxicology 2016, 59:167-182.
  • Provvisiero DP, Pivonello C, Muscogiuri G, Negri M, de Angelis C, Simeoli C, Pivonello R, Colao A. Influence of Bisphenol A on Type 2 Diabetes Mellitus. International Journal of Environmental Research and Public Health. 2016, 13(10). pii: E989.

Epigenetic effects of endocrine disrupting chemicals: Bisphenol A

Year 2016, Issue: 2, 186 - 203, 01.06.2016

Abstract

In the last decade, several comprehensive studies showed that epigenetic mechanisms could alter gene expressions and this phenomenon could lead to numerous diseases, particularly to cancer. Data indicate that some environmental contaminants, to which humans are widely exposed to, can cause epigenetic alterations. These alterations can play roles in the pathogenesis of some diseases. Bisphenol A BPA is one of the most abundant environmental contaminants and it is a chemical widely used in several fields, specifically in plastic industry. It is known that BPA has epigenetic effects in different periods of life and these effects have wide range of outcomes, like affecting fertility and cancer promotion. In this review, we will mainly focus on epigenetic mechanisms and the results of in vitro and in vivo studies, in which .the epigenetic effects of BPA were evaluated, will be presented.

References

  • Jiang Y, Bressler J, Beaudet LA: Epigenetics and human disease. Annual Reviews of Genetics 2004, 5:479-510.
  • Chatterjee R, Vinson C: CpG methylation recruits sequence specific transcription factors essential for tissue specific gene expression. Biochimica et Biophysica Acta. 2012, 1819(7):763-770.
  • Deaton AM, Bird A: CpG islands and the regulation of transcription. Genes and Development 2011, 25(10):1010-1022.
  • Kulis M, Queirós AC, Beekman R, Martín-Subero JI: Intragenic DNA methylation in transcriptional regulation, normal differentiation and cancer. Biochimica et Biophysica Acta 2013, 1829(11):1161-1174.
  • Meng H, Cao Y, Qin J, Song X, Zhang Q, Shi Y, Cao L: DNA methylation, its mediators and genome integrity. International Journal of Biological Sciences 2015, 11(5):604-617.
  • Erdmann A, Halby L, Fahy J, Arimondo PB: Targeting DNA methylation with small molecules: what’s next? Journal of Medicinal Chemistry 2015, 58(6):2569-2583.
  • Du J, Johnson LM, Jacobsen SE, Patel DJ: DNA methylation pathways and their crosstalk with histone methylation. Nature Reviews Molecular Cell Biology 2015, 16(9):519-532.
  • Dunn BK. Hypomethylation: one side of a larger picture. Annals of the New York Academy of Sciences. 2003, 983:28-42.
  • Wilson AS, Power BE, Molloy PL: DNA hypomethylation and human diseases. Biochim Biophys Acta. 2007, 1775(1):138-162.
  • Watanabe Y, Maekawa M: Methylation of DNA in cancer. Advances in Clinical Chemistry 2010, 52:145-167.
  • Zelic R, Fiano V, Grasso C, Zugna D, Pettersson A, Gillio-Tos A, Merletti F, Richiardi L: Global DNA hypomethylation in prostate cancer development and progression: a systematic review. Prostate Cancer and Prostatic Diseases 2015, 18(1):1-12.
  • Heichman KA, Warren JD: DNA methylation biomarkers and their utility for solid cancer diagnosticsClinical Chemistry and Laboratory Medicine 2012, 50(10):1707-1721.
  • Gokul G, Khosla S. DNA methylation and cancer. Subcell Biochem. 2013, 61:597-625.
  • Brennan K, Flanagan JM: Is there a link between genome-wide hypomethylation in blood and cancer risk? Cancer Prevention Research (Phila). 2012, 5(12):1345-1357.
  • Baba Y, Watanabe M, Baba H: Review of the alterations in DNA methylation in esophageal squamous cell carcinoma. Surgery Today 2013, 43(12):1355-1364.
  • Akhavan-Niaki H, Samadani AA: DNA methylation and cancer development: molecular mechanism. Cell Biochem Biophys. 2013, 67(2):501-513.
  • Woloszynska-Read A, Zhang W, Yu J, Link PA, Mhawech-Fauceglia P, Collamat G, Akers SN, Ostler KR, Godley LA, Odunsi K, Karpf AR: Coordinated cancer germline antigen promoter and global DNA hypomethylation in ovarian cancer: association with the BORIS/CTCF expression ratio and advanced stage. Clinical Cancer Research 2011, 17(8):2170-2180.
  • Lam K, Pan K, Linnekamp JF, Medema JP, Kandimalla R: DNA methylation based biomarkers in colorectal cancer: A systematic review. Biochimica et Biophysica Acta 2016, 1866(1):106-120.
  • Segre CV, Chiocca S: Regulating the regulators: The Post-Translational Code of Class I HDAC1 and HDAC2. Journal of Biomedicine and Biotechnology 2011, 2011:690848.
  • Moazed D, Bühler M, Buker SM, Colmenares SU, Gerace EL, Gerber SA, Hong EJ, Motamedi MR, Verdel A, Villén J, Gygi SP: Studies on the mechanism of RNAi-dependent heterochromatin assembly. Cold Spring Harbor Symposia on Quantitative Biology 2006, 71:461-471.
  • Agrawal N, Dasaradhi PV, Mohmmed A, Malhotra P, Bhatnagar RK, Mukherjee SK: RNA interference: biology, mechanism, and applications. Microbiology and Molecular Biology Reviews 2003, 67(4):657-685.
  • Egger G, Liang G, Aparicio A, Jones AP: Epigenetics in human disease and prospects for epigenetic therapy. Nature 2004, 429:457-463.
  • Ghildiyal M, Zamore PD: Small silencing RNAs: an expanding universe. Nature Reviews Genetics 2009, 10(2):94-108.
  • Pratt AJ, MacRae IJ: The RNA-induced silencing complex: a versatile gene-silencing machine. Journal of Biological Chemistry 2009, 284(27):17897-17901.
  • Kim YJ, Maizel A, Chen X: Traffic into silence: endomembranes and post-transcriptional RNA silencing. EMBO Journal 2014, 33(9):968-980.
  • Bajrami E, Spiroski M: Genomic Imprinting. Open Access Macedonian Journal of Medical Sciences 2016, 4(1):181-184.
  • Nicholls RD, Knepper JL: Genome organization, function, and imprinting in Prader-Willi and Angelman syndromes. Annu Rev Genomics Human Genetics 2001;2:153-175.
  • Jacob KJ, Robinson WP, Lefebvre L: Beckwith-Wiedemann and Silver-Russell syndromes: opposite developmental imbalances in imprinted regulators of placental function and embryonic growth. Clinşcal Genetics 2013, 84(4):326-334.
  • Marczak-Hałupka A, Kalina MA, Tańska A, Chrzanowska KH: Silver-Russell Syndrome - Part I: Clinical Characteristics and Genetic Background. Pediatric Endocrinology, Diabetes and Metabolism 2015, 20(3):101-106.
  • Levine MA: An update on the clinical and molecular characteristics of pseudohypoparathyroidism. Curr Opinions in Endocrinology, Diabetes and Obesity 2012, 19(6):443-451.
  • Herrera BM, Keildson S, Lindgren CM: Genetics and epigenetics of obesity. Maturitas. 2011, 69(1):41-49.
  • Pfeifer K: Mechanisms of Genomic Imprinting American Journal of Human Genetics, 2000, 67:777-877
  • Sleutels F, Barlow DP, Lyle R: The uniqueness of the imprinting mechanism. Current Opinions in Genetics and Development 2000,10:229-233.
  • Fundele RH, Surani MA, Allen ND. Consequences of genomic imprinting for fetal development. In: Reik W, Surani A (eds), Genomic imprinting, IRL/Oxford University Press, Oxford, New York, Tokyo 1997: 98–112.
  • Lintelmann J, Katayama A, Kurıhara N, Shore L, Wenzel A: Endocrine disrupters in the environment IUPAC Technical Report, Pure and Applied Chemistry 2003, 75(5):631-681.
  • Staples CA, Dorn PB, Klecka GM, O’Block ST, Harris LR: A review of the environmental fate, effects, and exposures of bisphenol A. Chemosphere 1998, 36(10):2149-2173.
  • Vom Saal FS, Nagel SC, Coe BL, Angle BM, Taylor JA. The estrogenic endocrine disrupting chemical bisphenol A (BPA) and obesity. Molecular and Cellular Endocrinology 2012, 354(1-2):74-84.
  • Johnson S, Saxena P, Sahu R: Leaching of Bisphenol A from Baby Bottles. Proceedings of the National Academy of Sciences, India Section B: Biological Sciences 2015, 85(1):131-135.
  • Olea N, Pulgar R, Pérez P, Olea-Serrano F, Rivas A, Novillo-Fertrell A, Pedraza V, Soto AM, Sonnenschein C: Estrogenicity of resin-based composites and sealants in dentistry. Environmental Health Perspectives 1996,104(3):298-305.
  • Vandenberg LN, Hunt PA, Myers JP, Vom Saal FS: Human exposures to bisphenol A: mismatches between data and assumptions. Reviews in Environmental Health 2013, 28(1):37-58.
  • Fenichel P, Chevalier N, Brucker-Davis F: Bisphenol A: an endocrine and metabolic disruptor. Annuals of Endocrinology (Paris) 2013, 74(3):211-220.
  • Batra T: Bisphenol-A in canned food products: is it really required? Archives of Industrial Hygiene and Toxicology 2011, 62(4):381-384.
  • Tyl RW, Myers CB, Marr MC, Thomas BF, Keimowitz AR, Brine DR, Veselica MM, Fail PA, Chang TY, Seely JC, Joiner RL, Butala JH, Dimond SS, Cagen SZ, Shiotsuka RN, Stropp GD, Waechter JM: Three-generation reproductive toxicity study of dietary bisphenol A in CD Sprague- Dawley rats. Toxicological Sciences 2002, 68(1):121-146.
  • Weng YI, Hsu PY, Liyanarachchi S, Liu J, Deatherage DE, Huang YW, Zuo T, Rodriguez B, Lin CH, Cheng AL, Huang TH: Epigenetic influences of low-dose bisphenol A in primary human breast epithelial cells. Toxicology and Applied Pharmacology 2010, 248(2):111-121.
  • Nagelkerke A, Sieuwerts AM, Bussink J, Sweep FC, Look MP, Foekens JA, Martens JW, Span PN: LAMP3 is involved in tamoxifen resistance in breast cancer cells through the modulation of autophagy. Endocrine-Related Cancer. 2014, 21(1):101-112.
  • Doherty LF, Bromer JG, Zhou Y, Aldad TS, Taylor HS: In utero exposure to diethylstilbestrol (DES) or bisphenol-A (BPA) increases EZH2 expression in the mammary gland: an epigenetic mechanism linking endocrine disruptors to breast cancer. Hormones and Cancer 2010, 1(3):146-155.
  • Bhan A, Hussain I, Ansari KI, Bobzean SA, Perrotti LI, Mandal SS. Histone methyltransferase EZH2 is transcriptionally induced by estradiol as well as estrogenic endocrine disruptors bisphenol-A and diethylstilbestrol. Journal of Molecular0 Biology 2014, 426(20):3426-3441.
  • Avissar-Whiting M, Veiga KR, Uhl KM, Maccani MA, Gagne LA, Moen EL, Marsit CJ: Bisphenol A exposure leads to specific microRNA alterations in placental cells. Reproductive Toxicology 2010, 29(4):401-406.
  • Cho H, Kim S, Park H.-W, Oh M-J, Yu S, Lee S, Park C, Han J, Oh J-H, Hwang S, Yoon S-J: A relationship between miRNA and gene expression in the mouse sertoli cell line after exposure to bisphenol A. BioChip Journal 2010, 4(1):75–81.
  • Wang T, Han J, Duan X, Xiong B, Cui XS, Kim NH, Liu HL, Sun SC: The toxic effects and possible mechanisms of Bisphenol A on oocyte maturation of porcine in vitro. Oncotarget 2016, 7(22):32554-32565.
  • Yaoi T, Itoh K, Nakamura K, Ogi H, Fujiwara Y, Fushiki S: Genome-wide analysis of epigenomic alterations in fetal mouse forebrain after exposure to low doses of bisphenol A. Biochemical and Biophysical Research Communications 2008, 376(3):563–567.
  • Dolinoy DC, Huang D, Jirtle RL: Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development. Proceedings of National Academecy of Sciences USA. 2007, 104(32):13056–13061.
  • Kochmanski J, Marchlewicz EH, Savidge M, Montrose L, Faulk C, Dolinoy DC: Longitudinal effects of developmental bisphenol A and variable diet exposures on epigenetic drift in mice. Reproductive Toxicology 2016, pii: S0890-6238(16)30308-2.
  • Manikkam M, Guerrero-Bosagna C, Tracey R, Haque MM, Skinner MK: Transgenerational actions of environmental compounds on reproductive disease and identification of epigenetic biomarkers of ancestral exposures. PLoS One. 2012, 7:e31901.
  • Doshi T, Mehta SS, Dighe V, Balasinor N, Vanage G: Hypermethylation of estrogen receptor promoter region in adult testis of rats exposed neonatally to bisphenol A. Toxicology. 2011;289(2- 3):74–82.
  • Mao Z, Xia W, Chang H, Huo W, Li Y, Xu S: Paternal BPA exposure in early life alters Igf2 epigenetic status in sperm and induces pancreatic impairment in rat offspring. Toxicology Letters 2015 238(3):30-38.
  • Ho SM, Tang WY: Belmonte de Frausto J, Prins GS: Developmental exposure to estradiol and bisphenol a increases susceptibility to prostate carcinogenesis and epigenetically regulates phosphodiesterase type 4 variant 4. Cancer Research 2006, 66(11):5624–5632.
  • Prins GS, Tang WY, Belmonte J, Ho SM: Developmental exposure to bisphenol a increases prostate cancer susceptibility in adult rats: Epigenetic mode of action is implicated. Fertility and Sterility 2008, 89:e41.
  • Cheong A, Zhang X, Cheung YY, Tang WY, Chen J, Ye SH, Medvedovic M, Leung YK, Prins GS, Ho SM: DNA methylome changes by estradiol benzoate and bisphenol A links early-life environmental exposures to prostate cancer risk. Epigenetics 2016. DOI: 10.1080/15592294.2016.1208891.
  • Weinhouse C, Sartor MA, Faulk C, Anderson OS, Sant KE, Harris C, Dolinoy DC: Epigenome- wide DNA methylation analysis implicates neuronal and inflammatory signaling pathways in adult murine hepatic tumorigenesis following perinatal exposure to bisphenol A. Environmental and Molecular Mutagenesis 2016, 57(6):435-446.
  • Deb P, Bhan A, Hussain I, Ansari KI, Bobzean SA, Pandita TK, Perrotti LI, Mandal SS: Endocrine disrupting chemical, bisphenol-A, induces breast cancer associated gene HOXB9 expression in vitro and in vivo. Gene 2016, 590(2):234-243.
  • Lamartiniere CA, Jenkins S, Betancourt AM, Wang J, Russo J: Exposure to the Endocrine Disruptor Bisphenol A Alters Susceptibility for Mammary Cancer. Hormone Molecular Biology and Clinical Investigation 2011, 5(2):45-52.
  • Bhan A, Hussain I, Ansari KI, Bobzean SA, Perrotti LI, Mandal SS: Bisphenol-A and diethylstilbestrol exposure induces the expression of breast cancer associated long noncoding RNA HOTAIR in vitro and in vivo. Journal Of Steroid Biochemistry And Molecular Biology 2014, 141:160-170.
  • Dhimolea E, Wadia PR, Murray TJ, Settles ML, Treitman JD, Sonnenschein C, Shioda T, Soto AM: Prenatal exposure to BPA alters the epigenome of the rat mammary gland and increases the propensity to neoplastic development. PLoS One 2014, 9(7):e99800.
  • Greathouse KL, Bredfeldt T, Everitt JI, Lin K, Berry T, Kannan K, Mittelstadt ML, Ho SM, Walker CL: Environmental estrogens differentially engage the histone methyltransferase EZH2 to increase risk of uterine tumorigenesis. Molecular Cancer Research 2012, 10(4):546-557.
  • Faulk C, Kim JH, Jones TR, McEachin RC, Nahar MS, Dolinoy DC, Sartor MA: Bisphenol A-associated alterations in genome-wide DNA methylation and gene expression patterns reveal sequence-dependent and non-monotonic effects in human fetal liver. Environmental Epigenetics 2015, 1(1). pii: dvv006.
  • Faulk C, Kim JH, Anderson OS, Nahar MS, Jones TR, Sartor MA, Dolinoy DC: Detection of differential DNA methylation in repetitive DNA of mice and humans perinatally exposed to bisphenol A. Epigenetics. 2016, 11(7):489-500.
  • Kundakovic M, Champagne FA: Epigenetic perspective on the developmental effects of bisphenol A. Brain Behavior and Immunity 2011, 25(6):1084–1093.
  • Jones BA, Shimell JJ, Watson NV. Pre- and postnatal bisphenol A treatment results in persistent deficits in the sexual behavior of male rats, but not female rats, in adulthood. Hormones and Behavior 2011, 59(2):246-251.
  • Prins GS, Tang WY, Belmonte J, Ho SM. Perinatal exposure to oestradiol and bisphenol A alters the prostate epigenome and increases susceptibility to carcinogenesis. Basic Clinical Pharmacology and Toxicology 2008, 102(2):134-138.
  • Yin N, Yao X, Qin Z, Wang YL, Faiola F: Assessment of Bisphenol A (BPA) neurotoxicity in vitro with mouse embryonic stem cells. Journal of Environmetal Sciences (China) 2015, 36:181-187.
  • Li J, Fu KZ, Vemula S, Le XC, Li XF. Studying developmental neurotoxic effects of bisphenol A (BPA) using embryonic stem cells. Journal of Environmetal Sciences (China) 2015, 1;36:173-177.
  • Hessel EV, Ezendam J, van Broekhuizen FA, Hakkert B, DeWitt J, Granum B, Guzylack L, Lawrence BP, Penninks A, Rooney AA, Piersma AH, van Loveren H. Assessment of recent developmental immunotoxicity studies with bisphenol A in the context of the 2015 EFSA TDI. Reproductive Toxicology 2016, 65:448-456.
  • Robinson L, Miller R. The Impact of Bisphenol A and Phthalates on Allergy, Asthma, and Immune Function: a Review of Latest Findings. Current Environmental Health Reports 2015, 2(4):379-387.
  • Ziv-Gal A, Flaws JA. Evidence for bisphenol A-induced female infertility: a review (2007-2016). Fertility and Sterility 2016, 106(4):827-856.
  • Seachrist DD, Bonk KW, Ho SM, Prins GS, Soto AM, Keri RA. A review of the carcinogenic potential of bisphenol A. Reproductive Toxicology 2016, 59:167-182.
  • Provvisiero DP, Pivonello C, Muscogiuri G, Negri M, de Angelis C, Simeoli C, Pivonello R, Colao A. Influence of Bisphenol A on Type 2 Diabetes Mellitus. International Journal of Environmental Research and Public Health. 2016, 13(10). pii: E989.
There are 77 citations in total.

Details

Primary Language Turkish
Journal Section Research Article
Authors

Pınar Erkekoğlu This is me

Belma Koçer-gümüşel This is me

Publication Date June 1, 2016
Published in Issue Year 2016 Issue: 2

Cite

Vancouver Erkekoğlu P, Koçer-gümüşel B. Epigenetik Etkileri Olan Endokrin Bozucu Kimyasal Maddeler: Bisfenol A. HUJPHARM. 2016(2):186-203.