5
Views
2
CrossRef citations to date
0
Altmetric
Review

The emerging role of epigenetics and miRNAs in endometriosis

, , &
Pages 431-450 | Published online: 10 Jan 2014

References

  • Koninckx PR, Meuleman C, Demeyere S, Lesaffre E, Cornillie FJ. Suggestive evidence that pelvic endometriosis is a progressive disease, whereas deeply infiltrating endometriosis is associated with pelvic pain. Fertil. Steril.55(4), 759–765 (1991).
  • Houston DE. Evidence for the risk of pelvic endometriosis by age, race and socioeconomic status. Epidemiol. Rev.6, 167–191 (1984).
  • Rothman KJ, Greenland S. Modern Epidemiology (2nd Edition). Lippincott–Raven, Philadelphia, PA, USA (1998).
  • Pardanani S, Barbieri RL. The gold standard for the surgical diagnosis of endometriosis: visual findings or biopsy results. J. Gynecol. Tech.4, 121–124 (1998).
  • Tanahatoe S, Hompes PG, Lambalk CB. Accuracy of diagnostic laparoscopy in the infertility work-up before intrauterine insemination. Fertil. Steril.79(2), 361–366 (2003).
  • Peterson EP, Behrman SJ. Laparoscopy of the infertile patient. Obstet. Gynecol.36, 363–370 (1970).
  • Hasson HM. Incidence of endometriosis in diagnostic laparoscopy. J. Reprod. Med.16(3), 135–138 (1976).
  • Pittaway DE. Diagnosis of endometriosis. Infertil. Reprod. Med. Clin. North. Am.3, 619–631 (1992).
  • Sampson JA. Peritoneal endometriosis due to the menstrual dissemination of endometrial tissue into the peritoneal cavity. Am. J. Obstet. Gynecol.12, 422 (1927).
  • Halme J, Hammond MG, Hulka JF, Raj SG, Talbert LM. Retrograde menstruation in healthy women and in patients with endometriosis. Obstet. Gynecol.64(2), 151–154 (1984).
  • Jimbo H, Hitomi Y, Yoshikawa H et al. Evidence for monoclonal expansion of epithelial cells in ovarian endometrial cysts. Am. J. Pathol.150(4), 1173–1178 (1997).
  • Fujii S. Secondary mullerian system and endometriosis. Am. J. Obstet. Gynecol.165(1), 219–225 (1991).
  • Mok-Lin EY, Wolfberg A, Hollinquist H, Laufer MR. Endometriosis in a Patient with Mayer–Rokitansky–Kuster–Hauser syndrome and complete uterine agenesis: evidence to support the theory of coelomic metaplasia. J. Pediatr. Adolesc. Gynecol.23(1), e35–e37 (2009).
  • Lebovic DI, Mueller MD, Taylor RN. Immunobiology of endometriosis. Fertil. Steril.75(1), 1–10 (2001).
  • Porpora MG, Ingelido AM, di Domenico A et al. Increased levels of polychlorobiphenyls in Italian women with endometriosis. Chemosphere63(8), 1361–1367 (2006).
  • Rier SE, Martin DC, Bowman RE, Dmowski WP, Becker JL. Endometriosis in rhesus monkeys (Macaca mulatta) following chronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Fundam. Appl. Toxicol.21(4), 433–441 (1993).
  • Cummings AM, Metcalf JL, Birnbaum L. Promotion of endometriosis by 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats and mice: time–dose dependence and species comparison. Toxicol. Appl. Pharmacol.138(1), 131–139 (1996).
  • Heilier JF, Donnez J, Lison D. Organochlorines and endometriosis: a mini-review. Chemosphere71(2), 203–210 (2008).
  • Guo SW. The link between exposure to dioxin and endometriosis: a critical reappraisal of primate data. Gynecol. Obstet. Invest.57(3), 157–173 (2004).
  • Matalliotakis IM, Arici A, Cakmak H, Goumenou AG, Koumantakis G, Mahutte NG. Familial aggregation of endometriosis in the Yale Series. Arch. Gynecol. Obstet.278(6), 507–511 (2008).
  • Treloar SA, Wicks J, Nyholt DR et al. Genomewide linkage study in 1,176 affected sister pair families identifies a significant susceptibility locus for endometriosis on chromosome 10q26. Am. J. Hum. Genet.77(3), 365–376 (2005).
  • Tempfer CB, Simoni M, Destenaves B, Fauser BC. Functional genetic polymorphisms and female reproductive disorders: part II – endometriosis. Hum. Reprod. Update15(1), 97–118 (2009).
  • Kitawaki J, Obayashi H, Ishihara H et al. Oestrogen receptor-α gene polymorphism is associated with endometriosis, adenomyosis and leiomyomata. Hum. Reprod.16(1), 51–55 (2001).
  • Arvanitis DA, Koumantakis GE, Goumenou AG, Matalliotakis IM, Koumantakis EE, Spandidos DA. CYP1A1, CYP19, and GSTM1 polymorphisms increase the risk of endometriosis. Fertil. Steril.79(Suppl. 1), 702–709 (2003).
  • Zervou S, Karteris E, Goumenou AG, Vatish M, Koumantakis EE, Hillhouse EW. The Glu298→Asp polymorphism of the endothelial nitric oxide synthase gene is associated with endometriosis. Fertil. Steril.80(6), 1524–1525 (2003).
  • Ammendola M, Gloria-Bottini F, Sesti F, Piccione E, Bottini E. Association of p53 codon 72 polymorphism with endometriosis. Fertil. Steril.90(2), 406–408 (2008).
  • Borghese B, Chiche JD, Vernerey D et al. Genetic polymorphisms of matrix metalloproteinase 12 and 13 genes are implicated in endometriosis progression. Hum. Reprod.23(5), 1207–1213 (2008).
  • Montgomery GW, Nyholt DR, Zhao ZZ et al. The search for genes contributing to endometriosis risk. Hum. Reprod. Update14(5), 447–457 (2008).
  • Di W, Guo SW. The search for genetic variants predisposing women to endometriosis. Curr. Opin. Obstet. Gynecol.19(4), 395–401 (2007).
  • Falconer H, D’Hooghe T, Fried G. Endometriosis and genetic polymorphisms. Obstet. Gynecol. Surv.62(9), 616–628 (2007).
  • Guo SW. Epigenetics of endometriosis. Mol. Hum. Reprod.15(10), 587–607 (2009).
  • Bestor TH. The DNA methyltransferases of mammals. Hum. Mol. Genet.9(16), 2395–2402 (2000).
  • Ehrlich M, Wang RY. 5-Methylcytosine in eukaryotic DNA. Science212(4501), 1350–1357 (1981).
  • Fuks F. DNA methylation and histone modifications: teaming up to silence genes. Curr. Opin. Genet. Dev.15(5), 490–495 (2005).
  • Robertson KD. DNA methylation and chromatin – unraveling the tangled web. Oncogene21(35), 5361–5379 (2002).
  • Peterson CL, Laniel MA. Histones and histone modifications. Curr. Biol.14(14), R546–R551 (2004).
  • Nakao M. Epigenetics: interaction of DNA methylation and chromatin. Gene278(1–2), 25–31 (2001).
  • Rodenhiser D, Mann M. Epigenetics and human disease: translating basic biology into clinical applications. CMAJ174(3), 341–348 (2006).
  • Ng HH, Bird A. DNA methylation and chromatin modification. Curr. Opin. Genet. Dev.9(2), 158–163 (1999).
  • Stein R, Sciaky-Gallili N, Razin A, Cedar H. Pattern of methylation of two genes coding for housekeeping functions. Proc. Natl Acad. Sci. USA80(9), 2422–2426 (1983).
  • Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell99(3), 247–257 (1999).
  • Hermann A, Schmitt S, Jeltsch A. The human Dnmt2 has residual DNA-(cytosine-C5) methyltransferase activity. J. Biol. Chem.278(34), 31717–31721 (2003).
  • Goll MG, Kirpekar F, Maggert KA et al. Methylation of tRNAAsp by the DNA methyltransferase homolog Dnmt2. Science311(5759), 395–398 (2006).
  • Suetake I, Shinozaki F, Miyagawa J, Takeshima H, Tajima S. DNMT3L stimulates the DNA methylation activity of Dnmt3a and Dnmt3b through a direct interaction. J. Biol. Chem.279(26), 27816–27823 (2004).
  • Gaudet F, Hodgson JG, Eden A et al. Induction of tumors in mice by genomic hypomethylation. Science300(5618), 489–492 (2003).
  • Rhee I, Bachman KE, Park BH et al. DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature416(6880), 552–556 (2002).
  • Wu Y, Strawn E, Basir Z, Halverson G, Guo SW. Aberrant expression of deoxyribonucleic acid methyltransferases DNMT1, DNMT3A, and DNMT3B in women with endometriosis. Fertil. Steril.87(1), 24–32 (2007).
  • Kao LC, Germeyer A, Tulac S et al. Expression profiling of endometrium from women with endometriosis reveals candidate genes for disease-based implantation failure and infertility. Endocrinology144(7), 2870–2881 (2003).
  • Burney RO, Talbi S, Hamilton AE et al. Gene expression analysis of endometrium reveals progesterone resistance and candidate susceptibility genes in women with endometriosis. Endocrinology148(8), 3814–3826 (2007).
  • Hull ML, Escareno CR, Godsland JM et al. Endometrial–peritoneal interactions during endometriotic lesion establishment. Am. J. Pathol.173(3), 700–715 (2008).
  • Eyster KM, Klinkova O, Kennedy V, Hansen KA. Whole genome deoxyribonucleic acid microarray analysis of gene expression in ectopic versus eutopic endometrium. Fertil. Steril.88(6), 1505–1533 (2007).
  • Eyster KM, Boles AL, Brannian JD, Hansen KA. DNA microarray analysis of gene expression markers of endometriosis. Fertil. Steril.77(1), 38–42 (2002).
  • Borghese B, Mondon F, Noel JC et al. Gene expression profile for ectopic versus eutopic endometrium provides new insights into endometriosis oncogenic potential. Mol. Endocrinol.22(11), 2557–2562 (2008).
  • Guo SW. Nuclear factor-κb (NF-κB): an unsuspected major culprit in the pathogenesis of endometriosis that is still at large? Gynecol. Obstet. Invest.63(2), 71–97 (2007).
  • Wren JD, Wu Y, Guo SW. A system-wide analysis of differentially expressed genes in ectopic and eutopic endometrium. Hum. Reprod.22(8), 2093–2102 (2007).
  • Jacinto FV, Ballestar E, Ropero S, Esteller M. Discovery of epigenetically silenced genes by methylated DNA immunoprecipitation in colon cancer cells. Cancer Res.67(24), 11481–11486 (2007).
  • Beliard A, Noel A, Foidart JM. Reduction of apoptosis and proliferation in endometriosis. Fertil. Steril.82(1), 80–85 (2004).
  • Braun DP, Ding J, Shaheen F, Willey JC, Rana N, Dmowski WP. Quantitative expression of apoptosis-regulating genes in endometrium from women with and without endometriosis. Fertil. Steril.87(2), 263–268 (2007).
  • Robertson KD, Keyomarsi K, Gonzales FA, Velicescu M, Jones PA. Differential mRNA expression of the human DNA methyltransferases (DNMTs) 1, 3a and 3b during the G(0)/G(1) to S phase transition in normal and tumor cells. Nucleic Acids Res.28(10), 2108–2113 (2000).
  • Issa JP, Kantarjian HM, Kirkpatrick P. Azacitidine. Nat. Rev. Drug Discov.4(4), 275–276 (2005).
  • Wu Y, Strawn E, Basir Z, Halverson G, Guo SW. Promoter hypermethylation of progesterone receptor isoform B (PR-B) in endometriosis. Epigenetics1(2), 106–111 (2006).
  • Gilbert J, Gore SD, Herman JG, Carducci MA. The clinical application of targeting cancer through histone acetylation and hypomethylation. Clin. Cancer Res.10(14), 4589–4596 (2004).
  • Izawa M, Harada T, Taniguchi F, Ohama Y, Takenaka Y, Terakawa N. An epigenetic disorder may cause aberrant expression of aromatase gene in endometriotic stromal cells. Fertil. Steril.89(Suppl. 5), 1390–1396 (2008).
  • Tsuchiya M, Nakao H, Katoh T et al. Association between endometriosis and genetic polymorphisms of the estradiol-synthesizing enzyme genes HSD17B1 and CYP19. Hum. Reprod.20(4), 974–978 (2005).
  • Hur SE, Lee S, Lee JY, Moon HS, Kim HL, Chung HW. Polymorphisms and haplotypes of the gene encoding the estrogen-metabolizing CYP19 gene in Korean women: no association with advanced-stage endometriosis. J. Hum. Genet.52(9), 703–711 (2007).
  • Kado N, Kitawaki J, Obayashi H et al. Association of the CYP17 gene and CYP19 gene polymorphisms with risk of endometriosis in Japanese women. Hum. Reprod.17(4), 897–902 (2002).
  • Guo SW. Association of endometriosis risk and genetic polymorphisms involving sex steroid biosynthesis and their receptors: a meta-analysis. Gynecol. Obstet. Invest.61(2), 90–105 (2006).
  • Colette S, Lousse JC, Defrere S et al. Absence of aromatase protein and mRNA expression in endometriosis. Hum. Reprod.24(9), 2133–2141 (2009).
  • Noble LS, Simpson ER, Johns A, Bulun SE. Aromatase expression in endometriosis. J. Clin. Endocrinol. Metab.81(1), 174–179 (1996).
  • Dassen H, Punyadeera C, Kamps R et al. Estrogen metabolizing enzymes in endometrium and endometriosis. Hum. Reprod.22(12), 3148–3158 (2007).
  • Nawathe A, Patwardhan S, Yates D, Harrison GR, Khan KS. Systematic review of the effects of aromatase inhibitors on pain associated with endometriosis. BJOG115(7), 818–822 (2008).
  • Attar E, Bulun SE. Aromatase inhibitors: the next generation of therapeutics for endometriosis? Fertil. Steril.85(5), 1307–1318 (2006).
  • Ferrero S, Camerini G, Seracchioli R, Ragni N, Venturini PL, Remorgida V. Letrozole combined with norethisterone acetate compared with norethisterone acetate alone in the treatment of pain symptoms caused by endometriosis. Hum. Reprod.24(12), 3033–3041 (2009).
  • Maia H Jr, Casoy J, Valente Filho J. Is aromatase expression in the endometrium the cause of endometriosis and related infertility? Gynecol. Endocrinol.25(4), 253–257 (2009).
  • Harada T, Yoshioka H, Yoshida S et al. Increased interleukin-6 levels in peritoneal fluid of infertile patients with active endometriosis. Am. J. Obstet. Gynecol.176(3), 593–597 (1997).
  • Eisermann J, Gast MJ, Pineda J, Odem RR, Collins JL. Tumor necrosis factor in peritoneal fluid of women undergoing laparoscopic surgery. Fertil. Steril.50(4), 573–579 (1988).
  • Zeitoun KM, Bulun SE. Aromatase: a key molecule in the pathophysiology of endometriosis and a therapeutic target. Fertil. Steril.72(6), 961–969 (1999).
  • Fechner S, Husen B, Thole H et al. Expression and regulation of estrogen-converting enzymes in ectopic human endometrial tissue. Fertil. Steril.88(Suppl. 4), 1029–1038 (2007).
  • Noble LS, Takayama K, Zeitoun KM et al. Prostaglandin E2 stimulates aromatase expression in endometriosis-derived stromal cells. J. Clin. Endocrinol. Metab.82(2), 600–606 (1997).
  • Huang JC, Dawood MY, Wu KK. Regulation of cyclooxygenase-2 gene in cultured endometrial stromal cells by sex steroids. Proceedings of: the 52nd Annual Meeting of the American Society for Reproductive Medicine. Boston, MA, USA, November 1996.
  • Zeitoun K, Takayama K, Michael MD, Bulun SE. Stimulation of aromatase P450 promoter (II) activity in endometriosis and its inhibition in endometrium are regulated by competitive binding of steroidogenic factor-1 and chicken ovalbumin upstream promoter transcription factor to the same cis-acting element. Mol. Endocrinol.13(2), 239–253 (1999).
  • Attar E, Tokunaga H, Imir G et al. Prostaglandin E2 via steroidogenic factor-1 coordinately regulates transcription of steroidogenic genes necessary for estrogen synthesis in endometriosis. J. Clin. Endocrinol. Metab.94(2), 623–631 (2009).
  • Xue Q, Lin Z, Yin P et al. Transcriptional activation of steroidogenic factor-1 by hypomethylation of the 5’ CpG island in endometriosis. J. Clin. Endocrinol. Metab.92(8), 3261–3267 (2007).
  • Chu S, Mamers P, Burger HG, Fuller PJ. Estrogen receptor isoform gene expression in ovarian stromal and epithelial tumors. J. Clin. Endocrinol. Metab.85(3), 1200–1205 (2000).
  • Parl FF. Estrogen receptor expression in breast cancer. In: Estrogens, Estrogen Receptor and Breast Cancer. IOS Press, Amsterdam, The Netherlands (2000).
  • Xue Q, Lin Z, Cheng YH et al. Promoter methylation regulates estrogen receptor 2 in human endometrium and endometriosis. Biol. Reprod.77(4), 681–687 (2007).
  • Olive DL, Pritts EA. Treatment of endometriosis. N. Engl. J. Med.345(4), 266–275 (2001).
  • Waller KG, Shaw RW. Gonadotropin-releasing hormone analogues for the treatment of endometriosis: long-term follow-up. Fertil. Steril.59(3), 511–515 (1993).
  • Vercellini P, Cortesi I, Crosignani PG. Progestins for symptomatic endometriosis: a critical analysis of the evidence. Fertil. Steril.68(3), 393–401 (1997).
  • Giangrande PH, McDonnell DP. The A and B isoforms of the human progesterone receptor: two functionally different transcription factors encoded by a single gene. Recent Prog. Horm. Res.54, 291–313; discussion 294–313 (1999).
  • Attia GR, Zeitoun K, Edwards D, Johns A, Carr BR, Bulun SE. Progesterone receptor isoform A but not B is expressed in endometriosis. J. Clin. Endocrinol. Metab.85(8), 2897–2902 (2000).
  • Krumlauf R. Hox genes in vertebrate development. Cell78(2), 191–201 (1994).
  • Taylor HS, Vanden Heuvel GB, Igarashi P. A conserved Hox axis in the mouse and human female reproductive system: late establishment and persistent adult expression of the Hoxa cluster genes. Biol. Reprod.57(6), 1338–1345 (1997).
  • Taylor HS, Bagot C, Kardana A, Olive D, Arici A. HOX gene expression is altered in the endometrium of women with endometriosis. Hum. Reprod.14(5), 1328–1331 (1999).
  • Browne H, Taylor H. HOXA10 expression in ectopic endometrial tissue. Fertil. Steril.85(5), 1386–1390 (2006).
  • Barbieri RL. Stenosis of the external cervical os: an association with endometriosis in women with chronic pelvic pain. Fertil. Steril.70(3), 571–573 (1998).
  • Liede A, Pal T, Mitchell M, Narod SA. Delineation of a new syndrome: clustering of pyloric stenosis, endometriosis, and breast cancer in two families. J. Med. Genet.37(10), 794–796 (2000).
  • Benson GV, Lim H, Paria BC, Satokata I, Dey SK, Maas RL. Mechanisms of reduced fertility in Hoxa-10 mutant mice: uterine homeosis and loss of maternal Hoxa-10 expression. Development122(9), 2687–2696 (1996).
  • Hsieh-Li HM, Witte DP, Weinstein M et al. Hoxa 11 structure, extensive antisense transcription, and function in male and female fertility. Development121(5), 1373–1385 (1995).
  • Kim JJ, Taylor HS, Lu Z et al. Altered expression of HOXA10 in endometriosis: potential role in decidualization. Mol. Hum. Reprod.13(5), 323–332 (2007).
  • Wu Y, Halverson G, Basir Z, Strawn E, Yan P, Guo SW. Aberrant methylation at HOXA10 may be responsible for its aberrant expression in the endometrium of patients with endometriosis. Am. J. Obstet. Gynecol.193(2), 371–380 (2005).
  • Lee B, Du H, Taylor HS. Experimental murine endometriosis induces DNA methylation and altered gene expression in eutopic endometrium. Biol. Reprod.80(1), 79–85 (2009).
  • Nabeshima H, Murakami T, Yoshinaga K, Sato K, Terada Y, Okamura K. Analysis of the clonality of ectopic glands in peritoneal endometriosis using laser microdissection. Fertil. Steril.80(5), 1144–1150 (2003).
  • Bromer JG, Wu J, Zhou Y, Taylor HS. Hypermethylation of homeobox A10 by in utero diethylstilbestrol exposure: an epigenetic mechanism for altered developmental programming. Endocrinology150(7), 3376–3382 (2009).
  • Lu Z, Hardt J, Kim JJ. Global analysis of genes regulated by HOXA10 in decidualization reveals a role in cell proliferation. Mol. Hum. Reprod.14(6), 357–366 (2008).
  • Nawroth F, Rahimi G, Nawroth C, Foth D, Ludwig M, Schmidt T. Is there an association between septate uterus and endometriosis? Hum. Reprod.21(2), 542–544 (2006).
  • Irwin JC, de las Fuentes L, Giudice LC. Growth factors and decidualization in vitro. Ann. NY Acad. Sci.734, 7–18 (1994).
  • Honda H, Barrueto FF, Gogusev J, Im DD, Morin PJ. Serial analysis of gene expression reveals differential expression between endometriosis and normal endometrium. Possible roles for AXL and SHC1 in the pathogenesis of endometriosis. Reprod. Biol. Endocrinol.6, 59 (2008).
  • Sbracia M, Zupi E, Alo P et al. Differential expression of IGF-I and IGF-II in eutopic and ectopic endometria of women with endometriosis and in women without endometriosis. Am. J. Reprod. Immunol.37(4), 326–329 (1997).
  • Feinberg AP, Cui H, Ohlsson R. DNA methylation and genomic imprinting: insights from cancer into epigenetic mechanisms. Semin. Cancer Biol.12(5), 389–398 (2002).
  • Ogawa O, Eccles MR, Szeto J et al. Relaxation of insulin-like growth factor II gene imprinting implicated in Wilms’ tumour. Nature362(6422), 749–751 (1993).
  • Matsuzaki S, Canis M, Darcha C, Dechelotte PJ, Pouly JL, Mage G. Expression of WT1 is downregulated in eutopic endometrium obtained during the midsecretory phase from patients with endometriosis. Fertil. Steril.86(3), 554–558 (2006).
  • Lee MP, Brandenburg S, Landes GM, Adams M, Miller G, Feinberg AP. Two novel genes in the center of the 11p15 imprinted domain escape genomic imprinting. Hum. Mol. Genet.8(4), 683–690 (1999).
  • Delaval K, Feil R. Epigenetic regulation of mammalian genomic imprinting. Curr. Opin. Genet. Dev.14(2), 188–195 (2004).
  • Bjornsson HT, Brown LJ, Fallin MD et al. Epigenetic specificity of loss of imprinting of the IGF2 gene in Wilms’ tumors. J. Natl Cancer Inst.99(16), 1270–1273 (2007).
  • Kozlov SV, Bogenpohl JW, Howell MP et al. The imprinted gene Magel2 regulates normal circadian output. Nat. Genet.39(10), 1266–1272 (2007).
  • Rainier S, Johnson LA, Dobry CJ, Ping AJ, Grundy PE, Feinberg AP. Relaxation of imprinted genes in human cancer. Nature362(6422), 747–749 (1993).
  • Ferguson JE 3rd, Wu Y, Smith K et al. ASB4 is a hydroxylation substrate of FIH and promotes vascular differentiation via an oxygen-dependent mechanism. Mol. Cell Biol.27(18), 6407–6419 (2007).
  • Yao I, Iida J, Nishimura W, Hata Y. Synaptic and nuclear localization of brain-enriched guanylate kinase-associated protein. J. Neurosci.22(13), 5354–5364 (2002).
  • Buntinx IM, Hennekam RC, Brouwer OF et al. Clinical profile of Angelman syndrome at different ages. Am. J. Med. Genet.56(2), 176–183 (1995).
  • Cassidy SB. Prader–Willi syndrome. Characteristics, management, and etiology. Ala. J. Med. Sci.24(2), 169–175 (1987).
  • Wilson MJ, Shivapurkar N, Poirier LA. Hypomethylation of hepatic nuclear DNA in rats fed with a carcinogenic methyl-deficient diet. Biochem. J.218(3), 987–990 (1984).
  • Ingrosso D, Cimmino A, Perna AF et al. Folate treatment and unbalanced methylation and changes of allelic expression induced by hyperhomocysteinaemia in patients with uraemia. Lancet361(9370), 1693–1699 (2003).
  • Locker J, Reddy TV, Lombardi B. DNA methylation and hepatocarcinogenesis in rats fed a choline-devoid diet. Carcinogenesis7(8), 1309–1312 (1986).
  • Asada K, Kotake Y, Asada R et al. LINE-1 hypomethylation in a choline-deficiency-induced liver cancer in rats: dependence on feeding period. J. Biomed. Biotechnol.2006(1), 17142 (2006).
  • Fjerbaek A, Knudsen UB. Endometriosis, dysmenorrhea and diet – what is the evidence? Eur. J. Obstet. Gynecol. Reprod. Biol.132(2), 140–147 (2007).
  • Zeisel SH. Importance of methyl donors during reproduction. Am. J. Clin. Nutr.89(2), 673S–677S (2009).
  • Mathers JC, McKay JA. Epigenetics – potential contribution to fetal programming. Adv. Exp. Med. Biol.646, 119–123 (2009).
  • Shukla SD, Velazquez J, French SW, Lu SC, Ticku MK, Zakhari S. Emerging role of epigenetics in the actions of alcohol. Alcohol Clin. Exp. Res.32(9), 1525–1534 (2008).
  • Reichman ME, Judd JT, Longcope C et al. Effects of alcohol consumption on plasma and urinary hormone concentrations in premenopausal women. J. Natl Cancer Inst.85(9), 722–727 (1993).
  • Mimura J, Fujii-Kuriyama Y. Functional role of AhR in the expression of toxic effects by TCDD. Biochim. Biophys. Acta.1619(3), 263–268 (2003).
  • Lebel G, Dodin S, Ayotte P, Marcoux S, Ferron LA, Dewailly E. Organochlorine exposure and the risk of endometriosis. Fertil. Steril.69(2), 221–228 (1998).
  • Tsukino H, Hanaoka T, Sasaki H et al. Associations between serum levels of selected organochlorine compounds and endometriosis in infertile Japanese women. Environ. Res.99(1), 118–125 (2005).
  • Heilier JF, Nackers F, Verougstraete V, Tonglet R, Lison D, Donnez J. Increased dioxin-like compounds in the serum of women with peritoneal endometriosis and deep endometriotic (adenomyotic) nodules. Fertil. Steril.84(2), 305–312 (2005).
  • Wu Q, Ohsako S, Ishimura R, Suzuki JS, Tohyama C. Exposure of mouse preimplantation embryos to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) alters the methylation status of imprinted genes H19 and Igf2. Biol. Reprod.70(6), 1790–1797 (2004).
  • Jenkins S, Rowell C, Wang J, Lamartiniere CA. Prenatal TCDD exposure predisposes for mammary cancer in rats. Reprod. Toxicol.23(3), 391–396 (2007).
  • Moffat ID, Boutros PC, Celius T, Linden J, Pohjanvirta R, Okey AB. microRNAs in adult rodent liver are refractory to dioxin treatment. Toxicol. Sci.99(2), 470–487 (2007).
  • Jin B, Kim G, Park DW, Ryu DY. Microarray analysis of gene regulation in the Hepa1c1c7 cell line following exposure to the DNA methylation inhibitor 5-aza-2’-deoxycytidine and 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol. In Vitro18(5), 659–664 (2004).
  • Missmer SA, Hankinson SE, Spiegelman D, Barbieri RL, Michels KB, Hunter DJ. In utero exposures and the incidence of endometriosis. Fertil. Steril.82(6), 1501–1508 (2004).
  • Li S, Hursting SD, Davis BJ, McLachlan JA, Barrett JC. Environmental exposure, DNA methylation, and gene regulation: lessons from diethylstilbesterol-induced cancers. Ann. NY Acad. Sci.983, 161–169 (2003).
  • Knight WA 3rd, Osborne CK, Yochmowitz MG, McGuire WL. Steroid hormone receptors in the management of human breast cancer. Ann. Clin. Res.12(5), 202–207 (1980).
  • McLachlan JA, Newbold RR, Bullock BC. Long-term effects on the female mouse genital tract associated with prenatal exposure to diethylstilbestrol. Cancer Res.40(11), 3988–3999 (1980).
  • Ruden DM, Xiao L, Garfinkel MD, Lu X. Hsp90 and environmental impacts on epigenetic states: a model for the trans-generational effects of diethylstibesterol on uterine development and cancer. Hum. Mol. Genet.14(Spec. 1), R149–R155 (2005).
  • Sato K, Fukata H, Kogo Y, Ohgane J, Shiota K, Mori C. Neonatal exposure to diethylstilbestrol alters the expression of DNA methyltransferases and methylation of genomic DNA in the epididymis of mice. Endocr. J.53(3), 331–337 (2006).
  • Aerts L, Van Assche FA. Is gestational diabetes an acquired condition? J. Dev. Physiol.1(3), 219–225 (1979).
  • Boloker J, Gertz SJ, Simmons RA. Gestational diabetes leads to the development of diabetes in adulthood in the rat. Diabetes51(5), 1499–1506 (2002).
  • Campbell JH, Perkins P. Transgenerational effects of drug and hormonal treatments in mammals: a review of observations and ideas. Prog. Brain Res.73, 535–553 (1988).
  • Newbold RR, Hanson RB, Jefferson WN, Bullock BC, Haseman J, McLachlan JA. Increased tumors but uncompromised fertility in the female descendants of mice exposed developmentally to diethylstilbestrol. Carcinogenesis19(9), 1655–1663 (1998).
  • Newbold RR, Padilla-Banks E, Jefferson WN. Adverse effects of the model environmental estrogen diethylstilbestrol are transmitted to subsequent generations. Endocrinology147(Suppl. 6), S11–S17 (2006).
  • Blatt J, Van Le L, Weiner T, Sailer S. Ovarian carcinoma in an adolescent with transgenerational exposure to diethylstilbestrol. J. Pediatr. Hematol. Oncol.25(8), 635–636 (2003).
  • Brouwers MM, Feitz WF, Roelofs LA, Kiemeney LA, de Gier RP, Roeleveld N. Hypospadias: a transgenerational effect of diethylstilbestrol? Hum. Reprod.21(3), 666–669 (2006).
  • Bruner-Tran KL, Yeaman GR, Eisenburg E, Osteen KG. Developmental dioxin exposure in mice alters uterine DNA methylation patterns and protein expression in adult animals and leads to an endometriosis-like phenotype. Presented at: 63rd Annual Meeting of the American Society of Reproductive Medicine. Washington, DC, USA (2007).
  • Morgan HD, Sutherland HG, Martin DI, Whitelaw E. Epigenetic inheritance at the agouti locus in the mouse. Nat. Genet.23(3), 314–318 (1999).
  • Waterland RA, Jirtle RL. Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol. Cell Biol.23(15), 5293–5300 (2003).
  • Waterland RA, Dolinoy DC, Lin JR, Smith CA, Shi X, Tahiliani KG. Maternal methyl supplements increase offspring DNA methylation at Axin Fused. Genesis44(9), 401–406 (2006).
  • Lillycrop KA, Phillips ES, Jackson AA, Hanson MA, Burdge GC. Dietary protein restriction of pregnant rats induces and folic acid supplementation prevents epigenetic modification of hepatic gene expression in the offspring. J. Nutr.135(6), 1382–1386 (2005).
  • Stein AD, Lumey LH. The relationship between maternal and offspring birth weights after maternal prenatal famine exposure: the Dutch Famine Birth Cohort Study. Hum. Biol.72(4), 641–654 (2000).
  • Painter RC, Roseboom TJ, Bleker OP. Prenatal exposure to the Dutch famine and disease in later life: an overview. Reprod. Toxicol.20(3), 345–352 (2005).
  • Kaati G, Bygren LO, Edvinsson S. Cardiovascular and diabetes mortality determined by nutrition during parents’ and grandparents’ slow growth period. Eur. J. Hum. Genet.10(11), 682–688 (2002).
  • Cotroneo MS, Lamartiniere CA. Pharmacologic, but not dietary, genistein supports endometriosis in a rat model. Toxicol. Sci.61(1), 68–75 (2001).
  • Yavuz E, Oktem M, Esinler I, Toru SA, Zeyneloglu HB. Genistein causes regression of endometriotic implants in the rat model. Fertil. Steril.88(Suppl. 4), 1129–1134 (2007).
  • Lim LP, Lau NC, Garrett-Engele P et al. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature433(7027), 769–773 (2005).
  • Teague EM, Print CG, Hull ML. The role of microRNAs in endometriosis and associated reproductive conditions. Hum. Reprod. Update16(2), 142–165 (2010).
  • Bernstein E, Kim SY, Carmell MA et al. Dicer is essential for mouse development. Nat. Genet.35(3), 215–217 (2003).
  • Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell116(2), 281–297 (2004).
  • Taganov KD, Boldin MP, Baltimore D. MicroRNAs and immunity: tiny players in a big field. Immunity26(2), 133–137 (2007).
  • Sonkoly E, Stahle M, Pivarcsi A. MicroRNAs and immunity: novel players in the regulation of normal immune function and inflammation. Semin. Cancer Biol.18(2), 131–140 (2008).
  • He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat. Rev. Genet.5(7), 522–531 (2004).
  • Sylvestre Y, De Guire V, Querido E et al. An E2F/miR-20a autoregulatory feedback loop. J. Biol. Chem.282(4), 2135–2143 (2007).
  • Li X, Carthew RW. A microRNA mediates EGF receptor signaling and promotes photoreceptor differentiation in the Drosophila eye. Cell123(7), 1267–1277 (2005).
  • Fazi F, Rosa A, Fatica A et al. A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPα regulates human granulopoiesis. Cell123(5), 819–831 (2005).
  • Yoo AS, Greenwald I. LIN-12/Notch activation leads to microRNA-mediated down-regulation of Vav in C. elegans. Science310(5752), 1330–1333 (2005).
  • Johnston RJ Jr, Chang S, Etchberger JF, Ortiz CO, Hobert O. MicroRNAs acting in a double-negative feedback loop to control a neuronal cell fate decision. Proc. Natl Acad. Sci. USA102(35), 12449–12454 (2005).
  • Bandres E, Agirre X, Bitarte N et al. Epigenetic regulation of microRNA expression in colorectal cancer. Int. J. Cancer125(11), 2737–2443 (2009).
  • Lee KH, Lotterman C, Karikari C et al. Epigenetic silencing of microRNA miR-107 regulates cyclin-dependent kinase 6 expression in pancreatic cancer. Pancreatology9(3), 293–301 (2009).
  • Roman-Gomez J, Agirre X, Jimenez-Velasco A et al. Epigenetic regulation of microRNAs in acute lymphoblastic leukemia. J. Clin. Oncol.27(8), 1316–1322 (2009).
  • Saito Y, Jones PA. Epigenetic activation of tumor suppressor microRNAs in human cancer cells. Cell Cycle5(19), 2220–2222 (2006).
  • Sonkoly E, Wei T, Janson PC et al. MicroRNAs: novel regulators involved in the pathogenesis of psoriasis? PLoS One2(7), e610 (2007).
  • Pan Q, Luo X, Toloubeydokhti T, Chegini N. The expression profile of micro-RNA in endometrium and endometriosis and the influence of ovarian steroids on their expression. Mol. Hum. Reprod.13(11), 797–806 (2007).
  • Ohlsson Teague EM, Van der Hoek KH, Van der Hoek MB et al. MicroRNA-regulated pathways associated with endometriosis. Mol. Endocrinol.23(2), 265–275 (2009).
  • Pan Q, Chegini N. MicroRNA signature and regulatory functions in the endometrium during normal and disease states. Semin. Reprod. Med.26(6), 479–493 (2008).
  • Wickramasinghe NS, Manavalan TT, Dougherty SM, Riggs KA, Li Y, Klinge CM. Estradiol downregulates miR-21 expression and increases miR-21 target gene expression in MCF-7 breast cancer cells. Nucleic Acids Res.37(8), 2584–2595 (2009).
  • Cohen A, Shmoish M, Levi L, Cheruti U, Levavi-Sivan B, Lubzens E. Alterations in micro-ribonucleic acid expression profiles reveal a novel pathway for estrogen regulation. Endocrinology149(4), 1687–1696 (2008).
  • Wu Y, Guo SW. Suppression of IL-1β-induced COX-2 expression by trichostatin A (TSA) in human endometrial stromal cells. Eur. J. Obstet. Gynecol. Reprod. Biol.135(1), 88–93 (2007).
  • Wilson TJ, Hertzog PJ, Angus D, Munnery L, Wood EC, Kola I. Decreased natural killer cell activity in endometriosis patients: relationship to disease pathogenesis. Fertil. Steril.62(5), 1086–1088 (1994).
  • Szyllo K, Tchorzewski H, Banasik M, Glowacka E, Lewkowicz P, Kamer-Bartosinska A. The involvement of T lymphocytes in the pathogenesis of endometriotic tissues overgrowth in women with endometriosis. Mediators Inflamm.12(3), 131–138 (2003).
  • Gallinelli A, Chiossi G, Giannella L, Marsella T, Genazzani AD, Volpe A. Different concentrations of interleukins in the peritoneal fluid of women with endometriosis: relationships with lymphocyte subsets. Gynecol. Endocrinol.18(3), 144–151 (2004).
  • Wu MY, Ho HN. The role of cytokines in endometriosis. Am. J. Reprod. Immunol.49(5), 285–296 (2003).
  • Mori H, Sawairi M, Nakagawa M, Itoh N, Wada K, Tamaya T. Expression of interleukin-1 (IL-1) β messenger ribonucleic acid (mRNA) and IL-1 receptor antagonist mRNA in peritoneal macrophages from patients with endometriosis. Fertil. Steril.57(3), 535–542 (1992).
  • Tseng JF, Ryan IP, Milam TD et al. Interleukin-6 secretion in vitro is upregulated in ectopic and eutopic endometrial stromal cells from women with endometriosis. J. Clin. Endocrinol. Metab.81(3), 1118–1122 (1996).
  • Akoum A, Lawson C, McColl S, Villeneuve M. Ectopic endometrial cells express high concentrations of interleukin (IL)-8 in vivo regardless of the menstrual cycle phase and respond to oestradiol by upregulating IL-1-induced IL-8 expression in vitro. Mol. Hum. Reprod.7(9), 859–866 (2001).
  • Klein NA, Pergola GM, Rao-Tekmal R, Dey TD, Schenken RS. Enhanced expression of resident leukocyte interferon γ mRNA in endometriosis. Am. J. Reprod. Immunol.30(2–3), 74–81 (1993).
  • Oosterlynck DJ, Meuleman C, Waer M, Koninckx PR. Transforming growth factor-beta activity is increased in peritoneal fluid from women with endometriosis. Obstet. Gynecol.83(2), 287–292 (1994).
  • Chen DB, Yang ZM, Hilsenrath R, Le SP, Harper MJ. Stimulation of prostaglandin (PG) F2 α and PGE2 release by tumour necrosis factor-α and interleukin-1 α in cultured human luteal phase endometrial cells. Hum. Reprod.10(10), 2773–2780 (1995).
  • Zhang RJ, Wild RA, Ojago JM. Effect of tumor necrosis factor-α on adhesion of human endometrial stromal cells to peritoneal mesothelial cells: an in vitro system. Fertil. Steril.59(6), 1196–1201 (1993).
  • Garcia-Velasco JA, Arici A. Interleukin-8 stimulates the adhesion of endometrial stromal cells to fibronectin. Fertil. Steril.72(2), 336–340 (1999).
  • Fakih H, Baggett B, Holtz G, Tsang KY, Lee JC, Williamson HO. Interleukin-1: a possible role in the infertility associated with endometriosis. Fertil. Steril.47(2), 213–217 (1987).
  • Watanabe H, Iwase M, Ohashi M, Nagumo M. Role of interleukin-8 secreted from human oral squamous cell carcinoma cell lines. Oral Oncol.38(7), 670–679 (2002).
  • Shifren JL, Tseng JF, Zaloudek CJ et al. Ovarian steroid regulation of vascular endothelial growth factor in the human endometrium: implications for angiogenesis during the menstrual cycle and in the pathogenesis of endometriosis. J. Clin. Endocrinol. Metab.81(8), 3112–3118 (1996).
  • Tan XJ, Lang JH, Liu DY, Shen K, Leng JH, Zhu L. Expression of vascular endothelial growth factor and thrombospondin-1 mRNA in patients with endometriosis. Fertil. Steril.78(1), 148–153 (2002).
  • Pupo-Nogueira A, de Oliveira RM, Petta CA, Podgaec S, Dias JA Jr, Abrao MS. Vascular endothelial growth factor concentrations in the serum and peritoneal fluid of women with endometriosis. Int. J. Gynaecol. Obstet.99(1), 33–37 (2007).
  • Ortiz DD. Chronic pelvic pain in women. Am. Fam. Physician.77(11), 1535–1542 (2008).
  • Cheong YC, Shelton JB, Laird SM et al. IL-1, IL-6 and TNF-α concentrations in the peritoneal fluid of women with pelvic adhesions. Hum. Reprod.17(1), 69–75 (2002).
  • Matarese G, De Placido G, Nikas Y, Alviggi C. Pathogenesis of endometriosis: natural immunity dysfunction or autoimmune disease? Trends Mol. Med.9(5), 223–228 (2003).
  • Waldman SA, Terzic A. MicroRNA signatures as diagnostic and therapeutic targets. Clin Chem, 54(6), 943–944 (2008).
  • Shimoni Y, Friedlander G, Hetzroni G et al. Regulation of gene expression by small non-coding RNAs: a quantitative view. Mol. Syst. Biol.3, 138 (2007).
  • Backdahl L, Bushell A, Beck S. Inflammatory signalling as mediator of epigenetic modulation in tissue-specific chronic inflammation. Int. J. Biochem. Cell Biol.41(1), 176–184 (2009).
  • Valinluck V, Wu W, Liu P, Neidigh JW, Sowers LC. Impact of cytosine 5-halogens on the interaction of DNA with restriction endonucleases and methyltransferase. Chem. Res. Toxicol.19(4), 556–562 (2006).
  • Izawa M, Taniguchi F, Uegaki T et al. Demethylation of a nonpromoter cytosine–phosphate–guanine island in the aromatase gene may cause the aberrant upregulation in endometriotic tissues. Fertil. Steril.95(1), 33–39 (2010).
  • Borghese B, Barbaux S, Mondon F et al. Research resource: genome-wide profiling of methylated promoters in endometriosis reveals a subtelomeric location of hypermethylation. Mol. Endocrinol.24(9), 1872–1885 (2010).
  • Bulmer JN, Jones RK, Searle RF. Intraepithelial leukocytes in endometriosis and adenomyosis: comparison of eutopic and ectopic endometrium with normal endometrium. Hum. Reprod.13(10), 2910–2915 (1998).
  • Wu Y, Guo SW. Peroxisome proliferator-activated receptor-γ and retinoid X receptor agonists synergistically suppress proliferation of immortalized endometrial stromal cells. Fertil. Steril.91(Suppl. 5), 2142–2147 (2009).
  • Lu Y, Nie J, Liu X, Zheng Y, Guo SW. Trichostatin A, a histone deacetylase inhibitor, reduces lesion growth and hyperalgesia in experimentally induced endometriosis in mice. Hum. Reprod.25(4), 1014–1025 (2010).
  • Sales KJ, Jabbour HN. Cyclooxygenase enzymes and prostaglandins in pathology of the endometrium. Reproduction126(5), 559–567 (2003).
  • Berner C, Aumuller E, Gnauck A, Nestelberger M, Just A, Haslberger AG. Epigenetic control of estrogen receptor expression and tumor suppressor genes is modulated by bioactive food compounds. Ann. Nutr. Metab.57(3–4), 183–189 (2010).
  • Li Y, Tollefsbol TO. Impact on DNA methylation in cancer prevention and therapy by bioactive dietary components. Curr. Med. Chem.17(20), 2141–2151 (2010).
  • Bulun SE, Zeitoun KM, Takayama K, Sasano H. Molecular basis for treating endometriosis with aromatase inhibitors. Hum. Reprod. Update6(5), 413–418 (2000).
  • Shaco-Levy R, Sharabi S, Benharroch D, Piura B, Sion-Vardy N. Matrix metalloproteinases 2 and 9, E-cadherin, and β-catenin expression in endometriosis, low-grade endometrial carcinoma and non-neoplastic eutopic endometrium. Eur. J. Obstet. Gynecol. Reprod. Biol.139(2), 226–232 (2008).
  • Wu HH, Wang NM, Lin CY, Tsai HD. Genetic alterations of HOXA10 and their effect on the severity of endometriosis in a Taiwanese population. Reprod. Biomed. Online16(3), 416–424 (2008).
  • Chegini N. Emerging role of MicroRNAs in reproductive medicine. Preface. Semin. Reprod. Med.26(6), 449–451 (2008).
  • Sotnikova NY, Antsiferova YS, Shokhina MN. Local epidermal growth factor production in women with endometriosis. Russ. J. Immunol.6(1), 55–60 (2001).

Website

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.