#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Genome-Wide DNA Methylation Analysis Predicts an Epigenetic Switch for GATA Factor Expression in Endometriosis


Women develop endometriosis when endometrial tissue with altered sensitivity to ovarian hormones grows outside the uterus. The persistent survival of these cells results in chronic pelvic pain and infertility. Although the origin of the disease remains a mystery, it only occurs in women and menstruating primates, suggesting that the unique evolution behind primate uterine development and menstruation are linked to the disease. Epigenetic defects affecting the uterine physiological response to ovarian hormones are also involved in endometriosis, and several genes implicated in disease progression are differentially methylated. Here we compared DNA methylation with gene expression in endometriosis using large-scale arrays. By comparing healthy and diseased cells treated with or without hormones to mimic part of the menstrual cycle, we uncovered many differentially methylated genes with defective expression in endometriosis that also regulate the hormone-dependent aspects of menstruation. In addition to expanding our understanding of how methylation affects endometriosis many fold, this also led us to propose an epigenetic switch that permits GATA6 expression in endometriosis instead of GATA2, and this switch promotes the aberrant expression of many of the genes seen in endometriosis. Our work provides novel unifying insight into the cause and development of endometriosis.


Vyšlo v časopise: Genome-Wide DNA Methylation Analysis Predicts an Epigenetic Switch for GATA Factor Expression in Endometriosis. PLoS Genet 10(3): e32767. doi:10.1371/journal.pgen.1004158
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004158

Souhrn

Women develop endometriosis when endometrial tissue with altered sensitivity to ovarian hormones grows outside the uterus. The persistent survival of these cells results in chronic pelvic pain and infertility. Although the origin of the disease remains a mystery, it only occurs in women and menstruating primates, suggesting that the unique evolution behind primate uterine development and menstruation are linked to the disease. Epigenetic defects affecting the uterine physiological response to ovarian hormones are also involved in endometriosis, and several genes implicated in disease progression are differentially methylated. Here we compared DNA methylation with gene expression in endometriosis using large-scale arrays. By comparing healthy and diseased cells treated with or without hormones to mimic part of the menstrual cycle, we uncovered many differentially methylated genes with defective expression in endometriosis that also regulate the hormone-dependent aspects of menstruation. In addition to expanding our understanding of how methylation affects endometriosis many fold, this also led us to propose an epigenetic switch that permits GATA6 expression in endometriosis instead of GATA2, and this switch promotes the aberrant expression of many of the genes seen in endometriosis. Our work provides novel unifying insight into the cause and development of endometriosis.


Zdroje

1. MahmoodTA, TempletonA (1991) Prevalence and genesis of endometriosis. Hum Reprod 6: 544–549.

2. GiudiceLC (2010) Clinical practice. Endometriosis. N Engl J Med 362: 2389–2398.

3. BulunSE (2009) Endometriosis. N Engl J Med 360: 268–279.

4. GuoSW (2009) Epigenetics of endometriosis. Mol Hum Reprod 15: 587–607.

5. BurneyRO, GiudiceLC (2012) Pathogenesis and pathophysiology of endometriosis. Fertil Steril 98: 511–519.

6. BricouA, BattRE, ChapronC (2008) Peritoneal fluid flow influences anatomical distribution of endometriotic lesions: why Sampson seems to be right. Eur J Obstet Gynecol Reprod Biol 138: 127–134.

7. Slayden OD (2013) Induced Endometriosis in Nonhuman Primates. Biol Reprod.

8. HalmeJ, HammondMG, HulkaJF, RajSG, TalbertLM (1984) Retrograde menstruation in healthy women and in patients with endometriosis. Obstet Gynecol 64: 151–154.

9. SignorilePG, BaldiF, BussaniR, ViceconteR, BulzomiP, et al. (2012) Embryologic origin of endometriosis: analysis of 101 human female fetuses. J Cell Physiol 227: 1653–1656.

10. EmeraD, RomeroR, WagnerG (2012) The evolution of menstruation: a new model for genetic assimilation: explaining molecular origins of maternal responses to fetal invasiveness. Bioessays 34: 26–35.

11. RamathalCY, BagchiIC, TaylorRN, BagchiMK (2010) Endometrial decidualization: of mice and men. Semin Reprod Med 28: 17–26.

12. BrennerRM, WestNB (1975) Hormonal regulation of the reproductive tract in female mammals. Annu Rev Physiol 37: 273–302.

13. GellersenB, BrosensIA, BrosensJJ (2007) Decidualization of the human endometrium: mechanisms, functions, and clinical perspectives. Semin Reprod Med 25: 445–453.

14. FinnCA (1998) Menstruation: a nonadaptive consequence of uterine evolution. Q Rev Biol 73: 163–173.

15. TabibzadehS (1996) The signals and molecular pathways involved in human menstruation, a unique process of tissue destruction and remodelling. Mol Hum Reprod 2: 77–92.

16. AghajanovaL, VelardeMC, GiudiceLC (2010) Altered gene expression profiling in endometrium: evidence for progesterone resistance. Semin Reprod Med 28: 51–58.

17. NasuK, YugeA, TsunoA, NishidaM, NaraharaH (2009) Involvement of resistance to apoptosis in the pathogenesis of endometriosis. Histol Histopathol 24: 1181–1192.

18. YinX, PavoneME, LuZ, WeiJ, KimJJ (2012) Increased activation of the PI3K/AKT pathway compromises decidualization of stromal cells from endometriosis. J Clin Endocrinol Metab 97: E35–43.

19. WetendorfM, DeMayoFJ (2012) The progesterone receptor regulates implantation, decidualization, and glandular development via a complex paracrine signaling network. Mol Cell Endocrinol 357: 108–118.

20. Giudice L, Evers JLH, Healy DL (2012) Endometriosis: science and practice. Chichester, West Sussex: Wiley-Blackwell. p. p.

21. WuY, StrawnE, BasirZ, HalversonG, GuoSW (2007) Aberrant expression of deoxyribonucleic acid methyltransferases DNMT1, DNMT3A, and DNMT3B in women with endometriosis. Fertil Steril 87: 24–32.

22. BuzzioOL, LuZ, MillerCD, UntermanTG, KimJJ (2006) FOXO1A differentially regulates genes of decidualization. Endocrinology 147: 3870–3876.

23. HeverA, RothRB, HeveziP, MarinME, AcostaJA, et al. (2007) Human endometriosis is associated with plasma cells and overexpression of B lymphocyte stimulator. Proc Natl Acad Sci U S A 104: 12451–12456.

24. PavoneME, DysonM, ReirstadS, PearsonE, IshikawaH, et al. (2011) Endometriosis expresses a molecular pattern consistent with decreased retinoid uptake, metabolism and action. Hum Reprod 26: 2157–2164.

25. LiQ, KannanA, DeMayoFJ, LydonJP, CookePS, et al. (2011) The antiproliferative action of progesterone in uterine epithelium is mediated by Hand2. Science 331: 912–916.

26. XueQ, ZhouYF, ZhuSN, BulunSE (2011) Hypermethylation of the CpG island spanning from exon II to intron III is associated with steroidogenic factor 1 expression in stromal cells of endometriosis. Reprod Sci 18: 1080–1084.

27. HoivikEA, BjanesoyTE, BakkeM (2013) Epigenetic regulation of the gene encoding steroidogenic factor-1. Mol Cell Endocrinol 371(1–2): 133–9.

28. CakmakH, TaylorHS (2010) Molecular mechanisms of treatment resistance in endometriosis: the role of progesterone-hox gene interactions. Semin Reprod Med 28: 69–74.

29. SzczepanskaM, WirstleinP, SkrzypczakJ, JagodzinskiPP (2012) Expression of HOXA11 in the mid-luteal endometrium from women with endometriosis-associated infertility. Reprod Biol Endocrinol 10: 1.

30. AnsariKI, HussainI, ShresthaB, KasiriS, MandalSS (2011) HOXC6 Is transcriptionally regulated via coordination of MLL histone methylase and estrogen receptor in an estrogen environment. J Mol Biol 411: 334–349.

31. EtcheverryA, AubryM, de TayracM, VauleonE, BonifaceR, et al. (2010) DNA methylation in glioblastoma: impact on gene expression and clinical outcome. BMC Genomics 11: 701.

32. KandimallaR, van TilborgAA, KompierLC, StumpelDJ, StamRW, et al. (2012) Genome-wide analysis of CpG island methylation in bladder cancer identified TBX2, TBX3, GATA2, and ZIC4 as pTa-specific prognostic markers. Eur Urol 61: 1245–1256.

33. van KaamKJ, DelvouxB, RomanoA, D'HoogheT, DunselmanGA, et al. (2011) Deoxyribonucleic acid methyltransferases and methyl-CpG-binding domain proteins in human endometrium and endometriosis. Fertil Steril 95: 1421–1427.

34. DingYB, LongCL, LiuXQ, ChenXM, GuoLR, et al. (2012) 5-aza-2′-deoxycytidine leads to reduced embryo implantation and reduced expression of DNA methyltransferases and essential endometrial genes. PLoS One 7: e45364.

35. FurstRW, MeyerHH, SchweizerG, UlbrichSE (2012) Is DNA methylation an epigenetic contribution to transcriptional regulation of the bovine endometrium during the estrous cycle and early pregnancy? Mol Cell Endocrinol 348: 67–77.

36. GaoF, MaX, RusieA, HemingwayJ, OstmannAB, et al. (2012) Epigenetic changes through DNA methylation contribute to uterine stromal cell decidualization. Endocrinology 153: 6078–6090.

37. GrimaldiG, ChristianM, QuenbyS, BrosensJJ (2012) Expression of epigenetic effectors in decidualizing human endometrial stromal cells. Mol Hum Reprod 18: 451–458.

38. YamagataY, AsadaH, TamuraI, LeeL, MaekawaR, et al. (2009) DNA methyltransferase expression in the human endometrium: down-regulation by progesterone and estrogen. Hum Reprod 24: 1126–1132.

39. NasuK, KawanoY, TsukamotoY, TakanoM, TakaiN, et al. (2011) Aberrant DNA methylation status of endometriosis: epigenetics as the pathogenesis, biomarker and therapeutic target. J Obstet Gynaecol Res 37: 683–695.

40. KimJJ, TaylorHS, LuZ, LadhaniO, HastingsJM, et al. (2007) Altered expression of HOXA10 in endometriosis: potential role in decidualization. Mol Hum Reprod 13: 323–332.

41. BorgheseB, BarbauxS, MondonF, SantulliP, PierreG, et al. (2010) Research resource: genome-wide profiling of methylated promoters in endometriosis reveals a subtelomeric location of hypermethylation. Mol Endocrinol 24: 1872–1885.

42. AkbasGE, TaylorHS (2004) HOXC and HOXD gene expression in human endometrium: lack of redundancy with HOXA paralogs. Biol Reprod 70: 39–45.

43. PaiAA, BellJT, MarioniJC, PritchardJK, GiladY (2011) A genome-wide study of DNA methylation patterns and gene expression levels in multiple human and chimpanzee tissues. PLoS Genet 7: e1001316.

44. RoesslerJ, AmmerpohlO, GutweinJ, HasemeierB, AnwarSL, et al. (2012) Quantitative cross-validation and content analysis of the 450 k DNA methylation array from Illumina, Inc. BMC Res Notes 5: 210.

45. RosenbloomKR, SloanCA, MalladiVS, DreszerTR, LearnedK, et al. (2013) ENCODE data in the UCSC Genome Browser: year 5 update. Nucleic Acids Res 41: D56–63.

46. RobertsonKD (2005) DNA methylation and human disease. Nat Rev Genet 6: 597–610.

47. DoiA, ParkIH, WenB, MurakamiP, AryeeMJ, et al. (2009) Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet 41: 1350–1353.

48. LongHK, SimsD, HegerA, BlackledgeNP, KutterC, et al. (2013) Epigenetic conservation at gene regulatory elements revealed by non-methylated DNA profiling in seven vertebrates. Elife 2: e00348.

49. Xue Q, Xu Y, Yang H, Zhang L, Shang J, et al. (2013) Methylation of a Novel CpG Island of Intron 1 Is Associated With Steroidogenic Factor 1 Expression in Endometriotic Stromal Cells. Reprod Sci [epub ahead of print].

50. BulunSE, UtsunomiyaH, LinZ, YinP, ChengYH, et al. (2009) Steroidogenic factor-1 and endometriosis. Mol Cell Endocrinol 300: 104–108.

51. MaunakeaAK, ChepelevI, CuiK, ZhaoK (2013) Intragenic DNA methylation modulates alternative splicing by recruiting MeCP2 to promote exon recognition. Cell Res 23(11): 1256–69.

52. WanJ, OliverVF, ZhuH, ZackDJ, QianJ, et al. (2013) Integrative analysis of tissue-specific methylation and alternative splicing identifies conserved transcription factor binding motifs. Nucleic Acids Res 41(18): 8503–14.

53. Jimenez-UsecheI, KeJ, TianY, ShimD, HowellSC, et al. (2013) DNA methylation regulated nucleosome dynamics. Sci Rep 3: 2121.

54. HanH, CortezCC, YangX, NicholsPW, JonesPA, et al. (2011) DNA methylation directly silences genes with non-CpG island promoters and establishes a nucleosome occupied promoter. Hum Mol Genet 20: 4299–4310.

55. VanderkraatsND, HikenJF, DeckerKF, EdwardsJR (2013) Discovering high-resolution patterns of differential DNA methylation that correlate with gene expression changes. Nucleic Acids Res 41: 6816–6827.

56. ZanattaA, RochaAM, CarvalhoFM, PereiraRM, TaylorHS, et al. (2010) The role of the Hoxa10/HOXA10 gene in the etiology of endometriosis and its related infertility: a review. J Assist Reprod Genet 27: 701–710.

57. IssaJP, OttavianoYL, CelanoP, HamiltonSR, DavidsonNE, et al. (1994) Methylation of the oestrogen receptor CpG island links ageing and neoplasia in human colon. Nat Genet 7: 536–540.

58. OttavianoYL, IssaJP, ParlFF, SmithHS, BaylinSB, et al. (1994) Methylation of the estrogen receptor gene CpG island marks loss of estrogen receptor expression in human breast cancer cells. Cancer Res 54: 2552–2555.

59. BulunSE, MonsavaisD, PavoneME, DysonM, XueQ, et al. (2012) Role of estrogen receptor-beta in endometriosis. Semin Reprod Med 30: 39–45.

60. HewetsonA, HendrixEC, MansharamaniM, LeeVH, ChiltonBS (2002) Identification of the RUSH consensus-binding site by cyclic amplification and selection of targets: demonstration that RUSH mediates the ability of prolactin to augment progesterone-dependent gene expression. Mol Endocrinol 16: 2101–2112.

61. HeYY, HeXJ, GuoPF, DuMR, ShaoJ, et al. (2012) The decidual stromal cells-secreted CCL2 induces and maintains decidual leukocytes into Th2 bias in human early pregnancy. Clin Immunol 145: 161–173.

62. InmanD, KawanaK, SchustD, LiningerR, YoungS (2008) Cyclic regulation of T-Bet and GATA-3 in human endometrium. Reprod Sci 15: 83–90.

63. RubelCA, FrancoHL, JeongJW, LydonJP, DeMayoFJ (2012) GATA2 is expressed at critical times in the mouse uterus during pregnancy. Gene Expr Patterns 12: 196–203.

64. PavoneME, ReierstadS, SunH, MiladM, BulunSE, et al. (2010) Altered retinoid uptake and action contributes to cell survival in endometriosis. J Clin Endocrinol Metab 95: E300–309.

65. ChlonTM, CrispinoJD (2012) Combinatorial regulation of tissue specification by GATA and FOG factors. Development 139: 3905–3916.

66. MolkentinJD (2000) The zinc finger-containing transcription factors GATA-4, -5, and -6. Ubiquitously expressed regulators of tissue-specific gene expression. J Biol Chem 275: 38949–38952.

67. MorriseyEE, IpHS, LuMM, ParmacekMS (1996) GATA-6: a zinc finger transcription factor that is expressed in multiple cell lineages derived from lateral mesoderm. Dev Biol 177: 309–322.

68. BrewerA, GoveC, DaviesA, McNultyC, BarrowD, et al. (1999) The human and mouse GATA-6 genes utilize two promoters and two initiation codons. J Biol Chem 274: 38004–38016.

69. YinF, HerringBP (2005) GATA-6 can act as a positive or negative regulator of smooth muscle-specific gene expression. J Biol Chem 280: 4745–4752.

70. AnafV, SimonP, FaytI, NoelJ (2000) Smooth muscles are frequent components of endometriotic lesions. Hum Reprod 15: 767–771.

71. Barcena de ArellanoML, GerickeJ, ReicheltU, OkuducuAF, EbertAD, et al. (2011) Immunohistochemical characterization of endometriosis-associated smooth muscle cells in human peritoneal endometriotic lesions. Hum Reprod 26: 2721–2730.

72. ChristensenS, VerhageHG, NowakG, de LanerolleP, FlemingS, et al. (1995) Smooth muscle myosin II and alpha smooth muscle actin expression in the baboon (Papio anubis) uterus is associated with glandular secretory activity and stromal cell transformation. Biol Reprod 53: 598–608.

73. KimJJ, JaffeRC, FazleabasAT (1999) Blastocyst invasion and the stromal response in primates. Hum Reprod 14 Suppl 245–55.

74. MieheU, Neumaier-WagnerP, KadyrovM, GoyalP, AlferJ, et al. (2005) Concerted upregulation of CLP36 and smooth muscle actin protein expression in human endometrium during decidualization. Cells Tissues Organs 179: 109–114.

75. LavoieHA, KingSR (2009) Transcriptional regulation of steroidogenic genes: STARD1, CYP11A1 and HSD3B. Exp Biol Med (Maywood) 234: 880–907.

76. BulunSE, FangZ, ImirG, GuratesB, TamuraM, et al. (2004) Aromatase and endometriosis. Semin Reprod Med 22: 45–50.

77. TsaiSJ, WuMH, LinCC, SunHS, ChenHM (2001) Regulation of steroidogenic acute regulatory protein expression and progesterone production in endometriotic stromal cells. J Clin Endocrinol Metab 86: 5765–5773.

78. BirdA (2002) DNA methylation patterns and epigenetic memory. Genes Dev 16: 6–21.

79. NobleLS, SimpsonER, JohnsA, BulunSE (1996) Aromatase expression in endometriosis. J Clin Endocrinol Metab 81: 174–179.

80. MayKE, Conduit-HulbertSA, VillarJ, KirtleyS, KennedySH, et al. (2010) Peripheral biomarkers of endometriosis: a systematic review. Hum Reprod Update 16: 651–674.

81. MayKE, VillarJ, KirtleyS, KennedySH, BeckerCM (2011) Endometrial alterations in endometriosis: a systematic review of putative biomarkers. Hum Reprod Update 17: 637–653.

82. Buck LouisGM, HedigerML, PetersonCM, CroughanM, SundaramR, et al. (2011) Incidence of endometriosis by study population and diagnostic method: the ENDO study. Fertil Steril 96(2): 360–5.

83. NnoahamKE, HummelshojL, WebsterP, d'HoogheT, de Cicco NardoneF, et al. (2011) Impact of endometriosis on quality of life and work productivity: a multicenter study across ten countries. Fertil Steril 96(2): 366–373.

84. LairdPW (2003) The power and the promise of DNA methylation markers. Nat Rev Cancer 3: 253–266.

85. RyanIP, SchriockED, TaylorRN (1994) Isolation, characterization, and comparison of human endometrial and endometriosis cells in vitro. J Clin Endocrinol Metab 78: 642–649.

86. NobleLS, TakayamaK, ZeitounKM, PutmanJM, JohnsDA, et al. (1997) Prostaglandin E2 stimulates aromatase expression in endometriosis-derived stromal cells. J Clin Endocrinol Metab 82: 600–606.

87. GrozdanovPN, StoccoDM (2012) Short RNA Molecules with High Binding Affinity to the KH Motif of A-Kinase Anchoring Protein 1 (AKAP1): Implications for the Regulation of Steroidogenesis. Mol Endocrinol 26: 2104–2117.

88. NavarroA, YinP, MonsivaisD, LinSM, DuP, et al. (2012) Genome-wide DNA methylation indicates silencing of tumor suppressor genes in uterine leiomyoma. PLoS One 7: e33284.

89. DysonMT, KowalewskiMP, MannaPR, StoccoDM (2009) The differential regulation of steroidogenic acute regulatory protein-mediated steroidogenesis by type I and type II PKA in MA-10 cells. Mol Cell Endocrinol 300: 94–103.

90. DysonMT, JonesJK, KowalewskiMP, MannaPR, AlonsoM, et al. (2008) Mitochondrial A-kinase anchoring protein 121 binds type II protein kinase A and enhances steroidogenic acute regulatory protein-mediated steroidogenesis in MA-10 mouse leydig tumor cells. Biol Reprod 78: 267–277.

91. GentlemanRC, CareyVJ, BatesDM, BolstadB, DettlingM, et al. (2004) Bioconductor: open software development for computational biology and bioinformatics. Genome Biol 5: R80.

92. DuP, KibbeWA, LinSM (2008) lumi: a pipeline for processing Illumina microarray. Bioinformatics 24: 1547–1548.

93. Gentleman R (2005) Bioinformatics and computational biology solutions using R and Bioconductor. New York: Springer Science+Business Media. xix, 473 p. p.

94. SmythGK (2004) Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 3: Article3.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 3
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#