The Pathogenesis of Endometriosis: Molecular and Cell Biology Insights

International Journal of Molecular Sciences - Tập 20 Số 22 - Trang 5615
Antonio Simone Laganà1, Simone Garzon1, Martin Götte2, Paola Viganò3, Massimo Piergiuseppe Franchi4, Fabio Ghezzi1, Dan C. Martin5,6
1Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Piazza Biroldi 1, 21100 Varese, Italy
2Department of Gynecology and Obstetrics, Münster University Hospital, D-48149 Münster, Germany
3Reproductive Sciences Laboratory, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Via Olgettina 60, 20136 Milan, Italy
4Department of Obstetrics and Gynecology, AOUI Verona, University of Verona, Piazzale Aristide Stefani 1, 37126 Verona, Italy
5School of Medicine, University of Tennessee Health Science Center, 910 Madison Ave, Memphis, TN 38163, USA
6Virginia Commonwealth University, 907 Floyd Ave, Richmond, VA 23284, USA

Tóm tắt

The etiopathogenesis of endometriosis is a multifactorial process resulting in a heterogeneous disease. Considering that endometriosis etiology and pathogenesis are still far from being fully elucidated, the current review aims to offer a comprehensive summary of the available evidence. We performed a narrative review synthesizing the findings of the English literature retrieved from computerized databases from inception to June 2019, using the Medical Subject Headings (MeSH) unique ID term “Endometriosis” (ID:D004715) with “Etiology” (ID:Q000209), “Immunology” (ID:Q000276), “Genetics” (ID:D005823) and “Epigenesis, Genetic” (ID:D044127). Endometriosis may origin from Müllerian or non-Müllerian stem cells including those from the endometrial basal layer, Müllerian remnants, bone marrow, or the peritoneum. The innate ability of endometrial stem cells to regenerate cyclically seems to play a key role, as well as the dysregulated hormonal pathways. The presence of such cells in the peritoneal cavity and what leads to the development of endometriosis is a complex process with a large number of interconnected factors, potentially both inherited and acquired. Genetic predisposition is complex and related to the combined action of several genes with limited influence. The epigenetic mechanisms control many of the processes involved in the immunologic, immunohistochemical, histological, and biological aberrations that characterize the eutopic and ectopic endometrium in affected patients. However, what triggers such alterations is not clear and may be both genetically and epigenetically inherited, or it may be acquired by the particular combination of several elements such as the persistent peritoneal menstrual reflux as well as exogenous factors. The heterogeneity of endometriosis and the different contexts in which it develops suggest that a single etiopathogenetic model is not sufficient to explain its complex pathobiology.

Từ khóa


Tài liệu tham khảo

Vercellini, 2014, Endometriosis: Pathogenesis and treatment, Nat. Rev. Endocrinol., 10, 261, 10.1038/nrendo.2013.255

Garai, 2006, Endometriosis: Harmful survival of an ectopic tissue, Front. Biosci. J. Virtual Libr., 11, 595, 10.2741/1821

Matalliotakis, 2017, The role of gene polymorphisms in endometriosis, Mol. Med. Rep., 16, 5881, 10.3892/mmr.2017.7398

Hsiao, 2017, Epigenetic regulation of the pathological process in endometriosis, Reprod. Med. Biol., 16, 314, 10.1002/rmb2.12047

Clement, 2007, The pathology of endometriosis: A survey of the many faces of a common disease emphasizing diagnostic pitfalls and unusual and newly appreciated aspects, Adv. Anat. Pathol., 14, 241, 10.1097/PAP.0b013e3180ca7d7b

Burney, 2012, Pathogenesis and pathophysiology of endometriosis, Fertil. Steril., 98, 511, 10.1016/j.fertnstert.2012.06.029

Vitale, 2017, Unus pro omnibus, omnes pro uno: A novel, evidence-based, unifying theory for the pathogenesis of endometriosis, Med. Hypotheses, 103, 10, 10.1016/j.mehy.2017.03.032

Rapisarda, 2017, Anxiety and depression in patients with endometriosis: Impact and management challenges, Int. J. Womens Health, 9, 323, 10.2147/IJWH.S119729

Condemi, 2015, Analysis of psychopathological comorbidity behind the common symptoms and signs of endometriosis, Eur. J. Obstet. Gynecol. Reprod. Biol., 194, 30, 10.1016/j.ejogrb.2015.08.015

Vitale, 2016, A Systematic Review of the Association Between Psychiatric Disturbances and Endometriosis, J. Obstet. Gynaecol. Can., 38, 1079, 10.1016/j.jogc.2016.09.008

Petrosino, 2017, Comment on “Risk of developing major depression and anxiety disorders among women with endometriosis: A longitudinal follow-up study”, J. Affect. Disord., 208, 672, 10.1016/j.jad.2016.07.016

Raffaelli, 2018, Mesenteric vascular and nerve sparing surgery in laparoscopic segmental intestinal resection for deep infiltrating endometriosis, Eur. J. Obstet. Gynecol. Reprod. Biol., 231, 214, 10.1016/j.ejogrb.2018.10.057

Baggio, 2015, Delivery and pregnancy outcome in women with bowel resection for deep endometriosis: A retrospective cohort study, Gynecol. Surg., 12, 279, 10.1007/s10397-015-0901-9

Parazzini, 2004, Endometriosis: Epidemiology and aetiological factors, Best Pract. Res. Clin. Obstet. Gynaecol., 18, 177, 10.1016/j.bpobgyn.2004.01.007

Hediger, 2011, Incidence of endometriosis by study population and diagnostic method: The ENDO study, Fertil. Steril., 96, 360, 10.1016/j.fertnstert.2011.05.087

Missmer, 2003, The epidemiology of endometriosis, Obstet. Gynecol. Clin. North Am., 30, 1, 10.1016/S0889-8545(02)00050-5

Evers, 1994, Endometriosis does not exist; all women have endometriosis, Hum. Reprod. Oxf. Engl., 9, 2206, 10.1093/oxfordjournals.humrep.a138421

Evers, 2013, Is adolescent endometriosis a progressive disease that needs to be diagnosed and treated?, Hum. Reprod. Oxf. Engl., 28, 2023, 10.1093/humrep/det298

Sampson, 1921, Perforating hemorrhage (chocolate) cysts of the ovary: Their importance and especially their relation to pelvic adenomas of endometrial type (“adenomyoma” of the uterus, rectovaginal septum, sigmoid, etc.), Arch. Surg., 3, 245, 10.1001/archsurg.1921.01110080003001

Sampson, 1927, Peritoneal endometriosis due to the menstrual dissemination of endometrial tissue into the peritoneal cavity, Am. J. Obstet. Gynecol., 14, 422, 10.1016/S0002-9378(15)30003-X

Sampson, 1940, The development of the implantation theory for the origin of peritoneal endometriosis, Am. J. Obstet. Gynecol., 40, 549, 10.1016/S0002-9378(40)91238-8

Nisolle, 1997, Peritoneal endometriosis, ovarian endometriosis, and adenomyotic nodules of the rectovaginal septum are three different entities, Fertil. Steril., 68, 585, 10.1016/S0015-0282(97)00191-X

Koninckx, 2012, Deep endometriosis: Definition, diagnosis, and treatment, Fertil. Steril., 98, 564, 10.1016/j.fertnstert.2012.07.1061

Koninckx, 1994, Is mild endometriosis a disease?: Is mild endometriosis a condition occurring intermittently in all women?, Hum. Reprod., 9, 2202, 10.1093/oxfordjournals.humrep.a138419

Donnez, 2003, Typical and subtle atypical presentations of endometriosis, Obstet. Gynecol. Clin. North Am., 30, 83, 10.1016/S0889-8545(02)00054-2

Waldeyer, 1870, “Die epithelialen Eierstockgeschwülste.” Ins besonders die Kystome (The epithelial ovarian tumors, especially the cystic tumors), Arch Gynäkol, 1, 252, 10.1007/BF01814007

1996, Theories on the pathogenesis of endometriosis, Hum. Reprod. Oxf. Engl., 11, 53, 10.1093/humrep/11.suppl_3.53

Miyazaki, 2018, Generation of Progesterone-Responsive Endometrial Stromal Fibroblasts from Human Induced Pluripotent Stem Cells: Role of the WNT/CTNNB1 Pathway, Stem Cell Rep., 11, 1136, 10.1016/j.stemcr.2018.10.002

Lauchlan, 1972, THE SECONDARY MÜLLERIAN SYSTEM, Obstet. Gynecol. Surv., 27, 133, 10.1097/00006254-197203000-00001

Batt, 2007, Müllerianosis, Histol. Histopathol., 22, 1161

Signorile, 2009, Ectopic endometrium in human foetuses is a common event and sustains the theory of müllerianosis in the pathogenesis of endometriosis, a disease that predisposes to cancer, J. Exp. Clin. Cancer Res., 28, 49, 10.1186/1756-9966-28-49

Nawroth, 2006, Is there an association between septate uterus and endometriosis?, Hum. Reprod., 21, 542, 10.1093/humrep/dei344

Wolfberg, 2010, Endometriosis in a Patient with Mayer-Rokitansky-Küster-Hauser Syndrome and Complete Uterine Agenesis: Evidence to Support the Theory of Coelomic Metaplasia, J. Pediatr. Adolesc. Gynecol., 23, e35, 10.1016/j.jpag.2009.02.010

Troncon, 2014, Endometriosis in a patient with mayer-rokitansky-küster-hauser syndrome, Case Rep. Obstet. Gynecol., 2014, 376231

Batt, 2003, Endometriosis from thelarche to midteens: Pathogenesis and prognosis, prevention and pedagogy, J. Pediatr. Adolesc. Gynecol., 16, 337, 10.1016/j.jpag.2003.09.008

Suginami, 1991, A reappraisal of the coelomic metaplasia theory by reviewing, endometriosis occurring in unusual sites and instances, Am. J. Obstet. Gynecol., 165, 214, 10.1016/0002-9378(91)90254-O

Rei, 2018, Endometriosis in a Man as a Rare Source of Abdominal Pain: A Case Report and Review of the Literature, Case Rep. Obstet. Gynecol., 2018, 2083121

Cullen, T.S. (1896). Adeno-myoma uteri diffusum benignum, Johns Hopkins Press.

Meyer, 1903, Über eine adenomatöse Wucherung der Serosa in einer Bauchnarbe, Zeitschr Geburtsh Gynakol, 49, 32

Novak, 1931, Pelvic endometriosis: Spontaneous rupture of endometrial cysts, with a report of three cases, Am. J. Obstet. Gynecol., 22, 826, 10.1016/S0002-9378(31)90752-7

Hu, 2019, Increased circulating miR-370-3p regulates steroidogenic factor 1 in endometriosis, Am. J. Physiol. Endocrinol. Metab., 316, E373, 10.1152/ajpendo.00244.2018

Meyer, 1919, Uber den Stand der Frage der Adenomyositis und Adenomyome im allgeneinen und insbesondere uber Adenomyositis seroepithelialis und Adenomyometritis sarcomatosa, Zentralbl Gynäkol, 36, 745

Reverter, 2019, Total Circulating Microparticle Levels After Laparoscopic Surgical Treatment for Endometrioma: A Pilot, Prospective, Randomized Study Comparing Stripping with CO2 Laser Vaporization, J. Minim. Invasive Gynecol., 26, 450, 10.1016/j.jmig.2018.05.014

Levander, 1955, The Pathogenesis of Endometriosis, Acta Obstet. Gynecol. Scand., 34, 366, 10.3109/00016345509158287

Merrill, 1966, Endometrial induction of endometriosis across Millipore filters, Am J Obstet Gynecol, 94, 780

Maniglio, 2018, Catamenial pneumothorax caused by thoracic endometriosis, Radiol. Case Rep., 13, 81, 10.1016/j.radcr.2017.09.003

Turan, 1995, Endometriosis in Association with Müllerian Anomalies, Gynecol. Obstet. Investig., 40, 261, 10.1159/000292349

Olive, 1987, Endometriosis and mullerian anomalies, Obstet. Gynecol., 69, 412

Javert, 1949, Pathogenesis of endometriosis based on endometrial homeoplasia, direct extension, exfoliation and implantation, lymphatic and hematogenous metastasis. Including five case reports of endometrial tissue in pelvic lymph nodes, Cancer, 2, 399, 10.1002/1097-0142(194905)2:3<399::AID-CNCR2820020304>3.0.CO;2-L

Halme, 1984, Retrograde menstruation in healthy women and in patients with endometriosis, Obstet. Gynecol., 64, 151

Kruitwagen, 1991, Endometrial epithelial cells in peritoneal fluid during the early follicular phase, Fertil. Steril., 55, 297, 10.1016/S0015-0282(16)54119-3

Koks, 1997, Evaluation of a menstrual cup to collect shed endometrium for in vitro studies, Fertil. Steril., 68, 560, 10.1016/S0015-0282(97)00250-1

Vercellini, 1998, Is cystic ovarian endometriosis an asymmetric disease?, Br. J. Obstet. Gynaecol., 105, 1018, 10.1111/j.1471-0528.1998.tb10267.x

Vercellini, 2007, Asymmetry in distribution of diaphragmatic endometriotic lesions: Evidence in favour of the menstrual reflux theory, Hum. Reprod., 22, 2359, 10.1093/humrep/dem224

Jenkins, 1986, Endometriosis: Pathogenetic implications of the anatomic distribution, Obstet. Gynecol., 67, 335

Martin, D.C. (2019, September 15). Laparoscopic Appearance of Endometriosis. Available online: https://www.danmartinmd.com/files/lae1988.pdf.

Roman, 2016, Bowel occult microscopic endometriosis in resection margins in deep colorectal endometriosis specimens has no impact on short-term postoperative outcomes, Fertil. Steril., 105, 423, 10.1016/j.fertnstert.2015.09.030

Badescu, 2016, Mapping of bowel occult microscopic endometriosis implants surrounding deep endometriosis nodules infiltrating the bowel, Fertil. Steril., 105, 430, 10.1016/j.fertnstert.2015.11.006

Mokshagundam, 2018, Rodent Models of Experimental Endometriosis: Identifying Mechanisms of Disease and Therapeutic Targets, Curr. Womens Health Rev., 14, 173, 10.2174/1573404813666170921162041

Garzon, 2018, Translational animal models for endometriosis research: A long and windy road, Ann. Transl. Med., 6, 431, 10.21037/atm.2018.08.24

Klemmt, 2018, Molecular and Cellular Pathogenesis of Endometriosis, Curr. Womens Health Rev., 14, 106, 10.2174/1573404813666170306163448

Ota, 1993, Expression of major histocompatibility complex class II antigen in endometriotic tissue in patients with endometriosis and adenomyosis, Fertil. Steril., 60, 834, 10.1016/S0015-0282(16)56284-0

Chung, 2001, Matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-3 mRNA expression in ectopic and eutopic endometrium in women with endometriosis: A rationale for endometriotic invasiveness, Fertil. Steril., 75, 152, 10.1016/S0015-0282(00)01670-8

Bonifacio, 2009, Metalloproteinases, vascular endothelial growth factor, and angiopoietin 1 and 2 in eutopic and ectopic endometrium, Fertil. Steril., 91, 2315, 10.1016/j.fertnstert.2008.03.079

Redwine, 2002, Was Sampson wrong?, Fertil. Steril., 78, 686, 10.1016/S0015-0282(02)03329-0

Suda, 2018, Clonal Expansion and Diversification of Cancer-Associated Mutations in Endometriosis and Normal Endometrium, Cell Rep., 24, 1777, 10.1016/j.celrep.2018.07.037

Leyendecker, 2009, The pathophysiology of endometriosis and adenomyosis: Tissue injury and repair, Arch. Gynecol. Obstet., 280, 529, 10.1007/s00404-009-1191-0

Young, 2017, The role of TGF-β in the pathophysiology of peritoneal endometriosis, Hum. Reprod. Update, 23, 548, 10.1093/humupd/dmx016

Maas, 2001, Development of endometriosis-like lesions after transplantation of human endometrial fragments onto the chick embryo chorioallantoic membrane, Hum. Reprod., 16, 627, 10.1093/humrep/16.4.627

Nap, 2003, Tissue integrity is essential for ectopic implantation of human endometrium in the chicken chorioallantoic membrane, Hum. Reprod., 18, 30, 10.1093/humrep/deg033

Winterhager, E. (2012). Role of Steroid Hormones: Estrogen and Endometriosis. Endometriosis: Science and Practice, John Wiley & Sons, Ltd.

Liang, 2018, Villainous role of estrogen in macrophage-nerve interaction in endometriosis, Reprod. Biol. Endocrinol., 16, 122, 10.1186/s12958-018-0441-z

Rier, 1995, Immunoresponsiveness in endometriosis: Implications of estrogenic toxicants, Environ. Health Perspect., 103, 151

Maruyama, 2008, Molecular and cellular mechanisms for differentiation and regeneration of the uterine endometrium, Endocr. J., 55, 795, 10.1507/endocrj.K08E-067

Ribeiro, 2019, Symptomatic endometriosis developing several years after menopause in the absence of increased circulating estrogen concentrations: A systematic review and seven case reports, Gynecol. Surg., 16, 3, 10.1186/s10397-019-1056-x

Kitawaki, 1997, Expression of aromatase cytochrome P450 protein and messenger ribonucleic acid in human endometriotic and adenomyotic tissues but not in normal endometrium, Biol. Reprod., 57, 514, 10.1095/biolreprod57.3.514

Noble, 1996, Aromatase expression in endometriosis, J. Clin. Endocrinol. Metab., 81, 174

Noble, 1997, Prostaglandin E2 stimulates aromatase expression in endometriosis-derived stromal cells, J. Clin. Endocrinol. Metab., 82, 600

Zeitoun, 1998, Deficient 17β-hydroxysteroid dehydrogenase type 2 expression in endometriosis: Failure to metabolize 17β-estradiol, J. Clin. Endocrinol. Metab., 83, 4474

TAKAHAsHI, 1989, Clinical usefulness of determination of estradiol level in the menstrual blood for patients with endometriosis, Nippon Sanka Fujinka Gakkai Zasshi, 41, 1849

Bulun, 2001, Role of aromatase in endometrial disease, J. Steroid Biochem. Mol. Biol., 79, 19, 10.1016/S0960-0760(01)00134-0

Wolf, 2008, Increased expression of the adult stem cell marker Musashi-1 in endometriosis and endometrial carcinoma, J. Pathol., 215, 317, 10.1002/path.2364

Valentijn, 2013, SSEA-1 isolates human endometrial basal glandular epithelial cells: Phenotypic and functional characterization and implications in the pathogenesis of endometriosis, Hum. Reprod. Oxf. Engl., 28, 2695, 10.1093/humrep/det285

Leyendecker, 2002, Endometriosis results from the dislocation of basal endometrium, Hum. Reprod., 17, 2725, 10.1093/humrep/17.10.2725

Agic, 2009, Apoptosis in endometriosis, Gynecol. Obstet. Investig., 68, 217, 10.1159/000235871

Zhao, 2015, Dual suppression of estrogenic and inflammatory activities for targeting of endometriosis, Sci. Transl. Med., 7, 271ra9, 10.1126/scitranslmed.3010626

Burns, 2012, Role of estrogen receptor signaling required for endometriosis-like lesion establishment in a mouse model, Endocrinology, 153, 3960, 10.1210/en.2012-1294

Han, 2015, Estrogen Receptor β Modulates Apoptosis Complexes and the Inflammasome to Drive the Pathogenesis of Endometriosis, Cell, 163, 960, 10.1016/j.cell.2015.10.034

Simmen, 2016, Reversal of Fortune: Estrogen Receptor-Beta in Endometriosis, J. Mol. Endocrinol., 57, F23, 10.1530/JME-16-0080

Hamilton, 2014, Estrogen hormone physiology: Reproductive findings from estrogen receptor mutant mice, Reprod. Biol., 14, 3, 10.1016/j.repbio.2013.12.002

Bulun, 2012, Role of Estrogen Receptor-β in Endometriosis, Semin. Reprod. Med., 30, 39, 10.1055/s-0031-1299596

Han, 2019, Genomic Function of Estrogen Receptor β in Endometriosis, Endocrinology, 160, 2495, 10.1210/en.2019-00442

McKinnon, 2018, Progesterone Resistance in Endometriosis: An Acquired Property?, Trends Endocrinol. Metab., 29, 535, 10.1016/j.tem.2018.05.006

Patel, 2017, Progesterone resistance in endometriosis: Origins, consequences and interventions, Acta Obstet. Gynecol. Scand., 96, 623, 10.1111/aogs.13156

Bulun, 2006, Progesterone resistance in endometriosis: Link to failure to metabolize estradiol, Mol. Cell. Endocrinol., 248, 94, 10.1016/j.mce.2005.11.041

Grandi, 2017, The association between progestins, nuclear receptors expression and inflammation in endometrial stromal cells from women with endometriosis, Gynecol. Endocrinol. Off. J. Int. Soc. Gynecol. Endocrinol., 33, 712, 10.1080/09513590.2017.1314458

Allport, 2001, Human labour is associated with nuclear factor-kappaB activity which mediates cyclo-oxygenase-2 expression and is involved with the “functional progesterone withdrawal”, Mol. Hum. Reprod., 7, 581, 10.1093/molehr/7.6.581

Cinar, 2009, Differential regulation of Akt phosphorylation in endometriosis, Reprod. Biomed. Online, 19, 864, 10.1016/j.rbmo.2009.10.001

Treloar, 2005, Association between polymorphisms in the progesterone receptor gene and endometriosis, Mol. Hum. Reprod., 11, 641, 10.1093/molehr/gah221

Ren, 2007, Down-regulation of the progesterone receptor by the methylation of progesterone receptor gene in endometrial cancer cells, Cancer Genet. Cytogenet., 175, 107, 10.1016/j.cancergencyto.2007.02.002

Teague, 2010, The role of microRNAs in endometriosis and associated reproductive conditions, Hum. Reprod. Update, 16, 142, 10.1093/humupd/dmp034

2015, Diagnostic potential of peritoneal fluid biomarkers of endometriosis, Expert Rev. Mol. Diagn., 15, 557, 10.1586/14737159.2015.1015994

Haney, 1981, Peritoneal fluid cell populations in infertility patients, Fertil. Steril., 35, 696, 10.1016/S0015-0282(16)45567-6

Hill, 1988, Characterization of leukocyte subpopulations in the peritoneal fluid of women with endometriosis, Fertil. Steril., 50, 216, 10.1016/S0015-0282(16)60062-6

Jones, 1998, Phenotypic and functional studies of leukocytes in human endometrium and endometriosis, Hum. Reprod. Update, 4, 702, 10.1093/humupd/4.5.702

Darrow, S.L., Vena, J.E., Batt, R.E., Zielezny, M.A., Michalek, A.M., and Selman, S. (1993). Menstrual cycle characteristics and the risk of endometriosis. Epidemiology, 135–142.

Vercellini, 1997, Menstrual characteristics in women with and without endometriosis, Obstet. Gynecol., 90, 264, 10.1016/S0029-7844(97)00235-4

Salamanca, 1995, Subendometrial contractility in menstrual phase visualized by transvaginal sonography in patients with endometriosis, Fertil. Steril., 64, 193, 10.1016/S0015-0282(16)57680-8

Sanfilippo, 1986, Endometriosis in association with uterine anomaly, Am. J. Obstet. Gynecol., 154, 39, 10.1016/0002-9378(86)90389-3

Oosterlynck, 1991, Women with endometriosis show a defect in natural killer activity resulting in a decreased cytotoxicity to autologous endometrium, Fertil. Steril., 56, 45, 10.1016/S0015-0282(16)54414-8

Triolo, 2016, Natural Killer T cell subsets in eutopic and ectopic endometrium: A fresh look to a busy corner, Arch. Gynecol. Obstet., 293, 941, 10.1007/s00404-015-4004-7

Semino, 1995, Role of major histocompatibility complex class I expression and natural killer-like T cells in the genetic control of endometriosis*, Fertil. Steril., 64, 909, 10.1016/S0015-0282(16)57901-1

Somigliana, 1996, Human endometrial stromal cells as a source of soluble intercellular adhesion molecule (ICAM)-1 molecules, Hum. Reprod. Oxf. Engl., 11, 1190, 10.1093/oxfordjournals.humrep.a019353

Gaffuri, 1998, Expression of intercellular adhesion molecule (ICAM)-1 mRNA and protein is enhanced in endometriosis versus endometrial stromal cells in culture, Mol. Hum. Reprod., 4, 1150, 10.1093/molehr/4.12.1150

Vetvicka, 2015, Immunological aspects of endometriosis: A review, Ann. Transl. Med., 3, 153

Riccio, 2018, Immunology of endometriosis, Best Pract. Res. Clin. Obstet. Gynaecol., 50, 39, 10.1016/j.bpobgyn.2018.01.010

Santulli, 2015, MAP kinases and the inflammatory signaling cascade as targets for the treatment of endometriosis?, Expert Opin. Ther. Targets, 19, 1465, 10.1517/14728222.2015.1090974

Balasch, 1997, Transferrin Receptor (CD71) Expression in Peritoneal Macrophages from Fertile and Infertile Women With and Without Endometriosis, Am. J. Reprod. Immunol., 38, 413, 10.1111/j.1600-0897.1997.tb00320.x

Etchepareborda, 1998, Functional and phenotypic alterations in peritoneal macrophages from patients with early and advanced endometriosis, Arch. Gynecol. Obstet., 261, 147, 10.1007/s004040050214

Halme, 1984, Accentuated cyclic activation of peritoneal macrophages in patients with endometriosis, Am. J. Obstet. Gynecol., 148, 85, 10.1016/S0002-9378(84)80037-X

McLaren, 1997, Immunolocalization of the apoptosis regulating proteins Bcl-2 and Bax in human endometrium and isolated peritoneal fluid macrophages in endometriosis, Hum. Reprod., 12, 146, 10.1093/humrep/12.1.146

Bacci, 2009, Macrophages Are Alternatively Activated in Patients with Endometriosis and Required for Growth and Vascularization of Lesions in a Mouse Model of Disease, Am. J. Pathol., 175, 547, 10.2353/ajpath.2009.081011

McLaren, 1997, Decreased levels of the potent regulator of monocyte/macrophage activation, interleukin-13, in the peritoneal fluid of patients with endometriosis, Hum. Reprod. Oxf. Engl., 12, 1307, 10.1093/humrep/12.6.1307

Schneider, 2013, Targeting of syndecan-1 by micro-ribonucleic acid miR-10b modulates invasiveness of endometriotic cells via dysregulation of the proteolytic milieu and interleukin-6 secretion, Fertil. Steril., 99, 871, 10.1016/j.fertnstert.2012.10.051

Joussen, 2002, Role of syndecan-1 in leukocyte-endothelial interactions in the ocular vasculature, Investig. Ophthalmol. Vis. Sci., 43, 1135

Averbeck, 2017, Syndecan-1 regulates dendritic cell migration in cutaneous hypersensitivity to haptens, Exp. Dermatol., 26, 1060, 10.1111/exd.13374

Pizzo, 2002, Behaviour of cytokine levels in serum and peritoneal fluid of women with endometriosis, Gynecol. Obstet. Investig., 54, 82, 10.1159/000067717

Malvezzi, 2017, “What do we know about regulatory T cells and endometriosis? A systematic review”, J. Reprod. Immunol., 120, 48, 10.1016/j.jri.2017.04.003

Riccio, 2017, The role of the B lymphocytes in endometriosis: A systematic review, J. Reprod. Immunol., 123, 29, 10.1016/j.jri.2017.09.001

Sattler, 1997, Structure of Bcl-xL-Bak peptide complex: Recognition between regulators of apoptosis, Science, 275, 983, 10.1126/science.275.5302.983

Tabibzadeh, 1995, Site and menstrual cycle-dependent expression of proteins of the tumour necrosis factor (TNF) receptor family, and BCL-2 oncoprotein and phase-specific production of TNF alpha in human endometrium, Hum. Reprod. Oxf. Engl., 10, 277, 10.1093/oxfordjournals.humrep.a135928

Taniguchi, 2011, Apoptosis and endometriosis, Front. Biosci. Elite Ed., 3, 648, 10.2741/e277

Gebel, 1998, Spontaneous apoptosis of endometrial tissue is impaired in women with endometriosis, Fertil. Steril., 69, 1042, 10.1016/S0015-0282(98)00073-9

Foidart, 2004, Reduction of apoptosis and proliferation in endometriosis, Fertil. Steril., 82, 80, 10.1016/j.fertnstert.2003.11.048

Lac, 2019, Iatrogenic endometriosis harbors somatic cancer-driver mutations, Hum. Reprod. Oxf. Engl., 34, 69, 10.1093/humrep/dey332

Vetvicka, 2016, Regulation of apoptotic pathways during endometriosis: From the molecular basis to the future perspectives, Arch. Gynecol. Obstet., 294, 897, 10.1007/s00404-016-4195-6

Sturlese, 2011, Dysregulation of the Fas/FasL system in mononuclear cells recovered from peritoneal fluid of women with endometriosis, J. Reprod. Immunol., 92, 74, 10.1016/j.jri.2011.08.005

Rutherford, E.J., Hill, A.D.K., and Hopkins, A.M. (2018). Adhesion in Physiological, Benign and Malignant Proliferative States of the Endometrium: Microenvironment and the Clinical Big Picture. Cells, 7.

Groothuis, 1999, Adhesion of human endometrial fragments to peritoneum in vitro, Fertil. Steril., 71, 1119, 10.1016/S0015-0282(99)00122-3

Koks, 1999, Adhesion of shed menstrual tissue in an in-vitro model using amnion and peritoneum: A light and electron microscopic study, Hum. Reprod. Oxf. Engl., 14, 816, 10.1093/humrep/14.3.816

Dunselman, 2001, The Mesothelium, Teflon or Velcro? Mesothelium in endometriosis pathogenesis, Hum. Reprod. Oxf. Engl., 16, 605, 10.1093/humrep/16.4.605

Canis, 2017, Trauma and endometriosis. A review. May we explain surgical phenotypes and natural history of the disease?, J. Gynecol. Obstet. Hum. Reprod., 46, 219, 10.1016/j.jogoh.2016.12.008

Koks, 2000, Menstruum induces changes in mesothelial cell morphology, Gynecol. Obstet. Investig., 50, 13, 10.1159/000010271

Groothuis, 2000, Morphological changes in mesothelial cells induced by shed menstrual endometrium in vitro are not primarily due to apoptosis or necrosis, Hum. Reprod. Oxf. Engl., 15, 1462, 10.1093/humrep/15.7.1462

Demir, 2004, Menstrual effluent induces epithelial-mesenchymal transitions in mesothelial cells, Hum. Reprod. Oxf. Engl., 19, 21, 10.1093/humrep/deh042

Bilyk, 2017, Epithelial-to-Mesenchymal Transition in the Female Reproductive Tract: From Normal Functioning to Disease Pathology, Front. Oncol., 5, 145, 10.3389/fonc.2017.00145

Yang, 2017, Epithelial-to-mesenchymal transition in the development of endometriosis, Oncotarget, 8, 41679, 10.18632/oncotarget.16472

Lessey, 1992, Integrin adhesion molecules in the human endometrium. Correlation with the normal and abnormal menstrual cycle, J. Clin. Investig., 90, 188, 10.1172/JCI115835

Lessey, 1998, Endometrial integrins and the establishment of uterine receptivity, Hum. Reprod., 13, 247, 10.1093/humrep/13.suppl_3.247

Tabibzadeh, 1992, Patterns of expression of integrin molecules in human endometrium throughout the menstrual cycle, Hum. Reprod., 7, 876, 10.1093/oxfordjournals.humrep.a137753

Dunselman, 1995, Expression of cadherins and integrins in human endometrium throughout the menstrual cycle**Supported in part by a research grant from Organon International B.V., Oss, The Netherlands, Fertil. Steril., 63, 1210, 10.1016/S0015-0282(16)57599-2

Lessey, 1994, Aberrant integrin expression in the endometrium of women with endometriosis, J. Clin. Endocrinol. Metab., 79, 643

Lessey, 2017, Endometrial receptivity in the eutopic endometrium of women with endometriosis: It is affected, and let me show you why, Fertil. Steril., 108, 19, 10.1016/j.fertnstert.2017.05.031

Germeyer, 2007, Expression of syndecans, cell-cell interaction regulating heparan sulfate proteoglycans, within the human endometrium and their regulation throughout the menstrual cycle, Fertil. Steril., 87, 657, 10.1016/j.fertnstert.2006.07.1526

Wilke, 2016, Syndecan-4 expression is upregulated in endometriosis and contributes to an invasive phenotype, Fertil. Steril., 106, 378, 10.1016/j.fertnstert.2016.03.032

Kotzian, 1999, The putative role of cell adhesion molecules in endometriosis: Can we learn from tumour metastasis?, Mol. Med. Today, 5, 304, 10.1016/S1357-4310(99)01497-5

Gaetje, 1997, Nonmalignant epithelial cells, potentially invasive in human endometriosis, lack the tumor suppressor molecule E-cadherin, Am. J. Pathol., 150, 461

Guilford, 1999, E-cadherin downregulation in cancer: Fuel on the fire?, Mol. Med. Today, 5, 172, 10.1016/S1357-4310(99)01461-6

Wijnhoven, 2000, E-cadherin-catenin cell-cell adhesion complex and human cancer, Br. J. Surg., 87, 992, 10.1046/j.1365-2168.2000.01513.x

Dunselman, 1994, Expression of integrins and E-cadherin in cells from menstrual effluent, endometrium, peritoneal fluid, peritoneum, and endometriosis, Fertil. Steril., 61, 85, 10.1016/S0015-0282(16)56457-7

Mashayekhi, 2015, Soluble CD44 concentration in the serum and peritoneal fluid samples of patients with different stages of endometriosis, Arch. Gynecol. Obstet., 292, 641, 10.1007/s00404-015-3654-9

Karousou, 2017, Roles and targeting of the HAS/hyaluronan/CD44 molecular system in cancer, Matrix Biol. J. Int. Soc. Matrix Biol., 59, 3, 10.1016/j.matbio.2016.10.001

Rodgers, 2011, Inhibition of CD44 N- and O-linked glycosylation decreases endometrial cell lines attachment to peritoneal mesothelial cells, Fertil. Steril., 95, 823, 10.1016/j.fertnstert.2010.09.005

Knudtson, 2016, Impaired Development of Early Endometriotic Lesions in CD44 Knockout Mice, Reprod. Sci. Thousand Oaks Calif, 23, 87, 10.1177/1933719115594022

Olivares, C.N., Alaniz, L.D., Menger, M.D., Barañao, R.I., Laschke, M.W., and Meresman, G.F. (2016). Inhibition of Hyaluronic Acid Synthesis Suppresses Angiogenesis in Developing Endometriotic Lesions. PloS ONE, 11.

Maksym, 2018, The bimodal role of matrix metalloproteinases and their inhibitors in etiology and pathogenesis of endometriosis, Mol. Med. Rep., 18, 3123

Murphy, 1994, Regulation of matrix metalloproteinase activity, Ann. N. Y. Acad. Sci., 732, 31, 10.1111/j.1749-6632.1994.tb24722.x

Nagase, 1997, Activation mechanisms of matrix metalloproteinases, Biol. Chem., 378, 151

Smigiel, 2017, Matrix Metalloproteinases and Leukocyte Activation, Prog. Mol. Biol. Transl. Sci., 147, 167, 10.1016/bs.pmbts.2017.01.003

Marbaix, 1995, The expression of interstitial collagenase in human endometrium is controlled by progesterone and by oestradiol and is related to menstruation, Biochem. J., 305, 1027, 10.1042/bj3051027

Hulboy, 1997, Matrix metalloproteinases as mediators of reproductive function, Mol. Hum. Reprod., 3, 27, 10.1093/molehr/3.1.27

Rodgers, 1994, Patterns of matrix metalloproteinase expression in cycling endometrium imply differential functions and regulation by steroid hormones, J. Clin. Investig., 94, 946, 10.1172/JCI117461

Bruner, 1999, Progesterone and transforming growth factor-β coordinately regulate suppression of endometrial matrix metalloproteinases in a model of experimental endometriosis, Steroids, 64, 648, 10.1016/S0039-128X(99)00048-3

Spuijbroek, 1992, Early endometriosis invades the extracellular matrix, Fertil. Steril., 58, 929, 10.1016/S0015-0282(16)55437-5

Osteen, 2002, Progesterone-Mediated Endometrial Maturation Limits Matrix Metalloproteinase (MMP) Expression in an Inflammatory-like Environment, Ann. N. Y. Acad. Sci., 955, 37, 10.1111/j.1749-6632.2002.tb02764.x

Salamonsen, 1996, Matrix metalloproteinases in normal menstruation, Hum. Reprod. Oxf. Engl., 11, 124, 10.1093/humrep/11.suppl_2.124

Salamonsen, 2000, Regulation of matrix metalloproteinases in human endometrium, Hum. Reprod. Oxf. Engl., 15, 112, 10.1093/humrep/15.suppl_3.112

Bruner, 1997, Suppression of matrix metalloproteinases inhibits establishment of ectopic lesions by human endometrium in nude mice, J. Clin. Investig., 99, 2851, 10.1172/JCI119478

Nap, 2004, Inhibiting MMP activity prevents the development of endometriosis in the chicken chorioallantoic membrane model, Hum. Reprod., 19, 2180, 10.1093/humrep/deh408

Osteen, 1996, Steroid and growth factor regulation of matrix metalloproteinase expression and endometriosis, Semin. Reprod. Endocrinol., 14, 247, 10.1055/s-2007-1016334

Sillem, 1998, Extracellular matrix remodelling in the endometrium and its possible relevance to the pathogenesis of endometriosis, Hum. Reprod. Update, 4, 730, 10.1093/humupd/4.5.730

Keisler, 1998, Tissue inhibitor of metalloproteinase-1 concentrations are attenuated in peritoneal fluid and sera of women with endometriosis and restored in sera by gonadotropin-releasing hormone agonist therapy, Fertil. Steril., 69, 1128, 10.1016/S0015-0282(98)00075-2

Cox, 2001, Differential regulation of matrix metalloproteinase-3 gene expression in endometriotic lesions compared with endometrium, Biol. Reprod., 65, 1297, 10.1095/biolreprod65.4.1297

Cox, 2002, Paracrine regulation of matrix metalloproteinase expression in endometriosis, Ann. N. Y. Acad. Sci., 955, 147, 10.1111/j.1749-6632.2002.tb02775.x

Pitsos, 2009, The role of matrix metalloproteinases in the pathogenesis of endometriosis, Reprod. Sci. Thousand Oaks Calif, 16, 717, 10.1177/1933719109333661

Zhou, 2005, The relevancy of the matrix metalloproteinase system to the pathophysiology of endometriosis, Front. Biosci. J. Virtual Libr., 10, 569, 10.2741/1552

Osteen, 2005, Reduced progesterone action during endometrial maturation: A potential risk factor for the development of endometriosis, Fertil. Steril., 83, 529, 10.1016/j.fertnstert.2004.11.026

Stamenkovic, 2003, Extracellular matrix remodelling: The role of matrix metalloproteinases, J. Pathol., 200, 448, 10.1002/path.1400

Maas, 1999, The chick embryo chorioallantoic membrane as a model to investigate the angiogenic properties of human endometrium, Gynecol. Obstet. Investig., 48, 108, 10.1159/000010150

Griffioen, 2000, Angiogenesis: Potentials for pharmacologic intervention in the treatment of cancer, cardiovascular diseases, and chronic inflammation, Pharmacol. Rev., 52, 237

McLaren, 2000, Vascular endothelial growth factor and endometriotic angiogenesis, Hum. Reprod. Update, 6, 45, 10.1093/humupd/6.1.45

Sharkey, 1993, Identification and localization of alternately spliced mRNAs for vascular endothelial growth factor in human uterus and estrogen regulation in endometrial carcinoma cell lines, Biol. Reprod., 48, 1120, 10.1095/biolreprod48.5.1120

Smith, 2001, Regulation of angiogenesis in the endometrium, Trends Endocrinol. Metab. TEM, 12, 147, 10.1016/S1043-2760(01)00379-4

Donnez, 1998, Vascular endothelial growth factor (VEGF) in endometriosis, Hum. Reprod. Oxf. Engl., 13, 1686, 10.1093/humrep/13.6.1686

Wingfield, 1995, Cell proliferation is increased in the endometrium of women with endometriosis, Fertil. Steril., 64, 340, 10.1016/S0015-0282(16)57733-4

Bourlev, 2006, The relationship between microvessel density, proliferative activity and expression of vascular endothelial growth factor-A and its receptors in eutopic endometrium and endometriotic lesions, Reproduction, 132, 501, 10.1530/rep.1.01110

Matsuzaki, 2001, Immunohistochemical analysis of the role of angiogenic status in the vasculature of peritoneal endometriosis, Fertil. Steril., 76, 712, 10.1016/S0015-0282(01)02003-9

Hull, 2003, Antiangiogenic agents are effective inhibitors of endometriosis, J. Clin. Endocrinol. Metab., 88, 2889, 10.1210/jc.2002-021912

Nap, 2005, Angiostatic agents prevent the development of endometriosis-like lesions in the chicken chorioallantoic membrane, Fertil. Steril., 83, 793, 10.1016/j.fertnstert.2004.06.080

Nap, 2004, Antiangiogenesis therapy for endometriosis, J. Clin. Endocrinol. Metab., 89, 1089, 10.1210/jc.2003-031406

Donnez, 2008, Antiangiogenic and vascular-disrupting agents in endometriosis: Pitfalls and promises, Mol. Hum. Reprod., 14, 259, 10.1093/molehr/gan019

Becker, 2007, Angiogenesis and antiangiogenic therapy in endometriosis, Microvasc. Res., 74, 121, 10.1016/j.mvr.2007.04.008

May, 2008, Endometriosis and angiogenesis, Minerva Ginecol., 60, 245

Taylor, 2009, Mechanistic and Therapeutic Implications of Angiogenesis in Endometriosis, Reprod. Sci. Thousand Oaks Calif, 16, 140, 10.1177/1933719108324893

Gerstner, 2018, Notch signaling controls sprouting angiogenesis of endometriotic lesions, Angiogenesis, 21, 37, 10.1007/s10456-017-9580-7

Maruyama, 2012, Stem cell theory for the pathogenesis of endometriosis, Front. Biosci. Elite Ed., 4, 2754, 10.2741/e589

Gargett, 2010, Adult stem cells in the endometrium, Mol. Hum. Reprod., 16, 818, 10.1093/molehr/gaq061

Taylor, 2004, Endometrial cells derived from donor stem cells in bone marrow transplant recipients, JAMA, 292, 81, 10.1001/jama.292.1.81

Masuda, H., Matsuzaki, Y., Hiratsu, E., Ono, M., Nagashima, T., Kajitani, T., Arase, T., Oda, H., Uchida, H., and Asada, H. (2010). Stem Cell-Like Properties of the Endometrial Side Population: Implication in Endometrial Regeneration. PLoS ONE, 5.

Wu, 2003, Resolution of clonal origins for endometriotic lesions using laser capture microdissection and the human androgen receptor (HUMARA) assay, Fertil. Steril., 79, 710, 10.1016/S0015-0282(02)04821-5

Nabeshima, 2003, Analysis of the clonality of ectopic glands in peritoneal endometriosis using laser microdissection, Fertil. Steril., 80, 1144, 10.1016/S0015-0282(03)01181-6

Mayr, 2003, Does endometriosis really have premalignant potential? A clonal analysis of laser-microdissected tissue, FASEB J., 17, 693, 10.1096/fj.02-0562fje

Maruyama, 2010, Human uterine stem/progenitor cells: Their possible role in uterine physiology and pathology, Reprod. Camb. Engl., 140, 11, 10.1530/REP-09-0438

Wang, Y., Sacchetti, A., van Dijk, M.R., van der Zee, M., van der Horst, P.H., Joosten, R., Burger, C.W., Grootegoed, J.A., Blok, L.J., and Fodde, R. (2012). Identification of Quiescent, Stem-Like Cells in the Distal Female Reproductive Tract. PLOS ONE, 7.

Salmeri, 2018, Stem Cell Trafficking During Endometriosis: May Epigenetics Play a Pivotal Role?, Reprod. Sci. Thousand Oaks Calif, 25, 978, 10.1177/1933719116687661

Dahlhues, 2018, The endometrial stem cell markers notch-1 and numb are associated with endometriosis, Reprod. Biomed. Online, 36, 294, 10.1016/j.rbmo.2017.11.010

Wolf, 2011, Aberrant expression of the pluripotency marker SOX-2 in endometriosis, Fertil. Steril., 95, 338, 10.1016/j.fertnstert.2010.08.006

Adammek, 2013, MicroRNA miR-145 inhibits proliferation, invasiveness, and stem cell phenotype of an in vitro endometriosis model by targeting multiple cytoskeletal elements and pluripotency factors, Fertil. Steril., 99, 1346, 10.1016/j.fertnstert.2012.11.055

Eggers, 2016, microRNA miR-200b affects proliferation, invasiveness and stemness of endometriotic cells by targeting ZEB1, ZEB2 and KLF4, Reprod. Biomed. Online, 32, 434, 10.1016/j.rbmo.2015.12.013

Sapkota, 2017, Meta-analysis identifies five novel loci associated with endometriosis highlighting key genes involved in hormone metabolism, Nat. Commun., 8, 15539, 10.1038/ncomms15539

Bouaziz, 2018, How Artificial Intelligence Can Improve Our Understanding of the Genes Associated with Endometriosis: Natural Language Processing of the PubMed Database, BioMed Res. Int., 2018, 6217812, 10.1155/2018/6217812

Gajbhiye, 2018, Complex genetics of female fertility, NPJ Genomic Med., 3, 29, 10.1038/s41525-018-0068-1

Kennedy, 1998, The genetics of endometriosis, J. Reprod. Med., 43, 263

Simpson, 2002, Heritability and molecular genetic studies of endometriosis, Ann. N. Y. Acad. Sci., 955, 239, 10.1111/j.1749-6632.2002.tb02785.x

Stefansson, 2002, Genetic factors contribute to the risk of developing endometriosis, Hum. Reprod. Oxf. Engl., 17, 555, 10.1093/humrep/17.3.555

Treloar, 2002, The International Endogene Study: A collection of families for genetic research in endometriosis, Fertil. Steril., 78, 679, 10.1016/S0015-0282(02)03341-1

Zondervan, 2001, The genetic basis of endometriosis, Curr. Opin. Obstet. Gynecol., 13, 309, 10.1097/00001703-200106000-00011

Zondervan, 2004, Familial aggregation of endometriosis in a large pedigree of rhesus macaques, Hum. Reprod. Oxf. Engl., 19, 448, 10.1093/humrep/deh052

Kennedy, 1995, Familial endometriosis, J. Assist. Reprod. Genet., 12, 32, 10.1007/BF02214126

Hadfield, 1997, Endometriosis in monozygotic twins, Fertil. Steril., 68, 941, 10.1016/S0015-0282(97)00359-2

Moen, 1994, Endometriosis in monozygotic twins, Acta Obstet. Gynecol. Scand., 73, 59, 10.3109/00016349409013396

Treloar, 1999, Genetic influences on endometriosis in an Australian twin sample, Fertil. Steril., 71, 701, 10.1016/S0015-0282(98)00540-8

Di, 2007, The search for genetic variants predisposing women to endometriosis, Curr. Opin. Obstet. Gynecol., 19, 395, 10.1097/GCO.0b013e328235a5b4

Falconer, 2007, Endometriosis and genetic polymorphisms, Obstet. Gynecol. Surv., 62, 616, 10.1097/01.ogx.0000279293.60436.60

Guo, 2005, Glutathione S-transferases M1/T1 gene polymorphisms and endometriosis: A meta-analysis of genetic association studies, Mol. Hum. Reprod., 11, 729, 10.1093/molehr/gah206

Guo, 2006, The association of endometriosis risk and genetic polymorphisms involving dioxin detoxification enzymes: A systematic review, Eur. J. Obstet. Gynecol. Reprod. Biol., 124, 134, 10.1016/j.ejogrb.2005.10.002

Guo, 2006, Association of endometriosis risk and genetic polymorphisms involving sex steroid biosynthesis and their receptors: A meta-analysis, Gynecol. Obstet. Investig., 61, 90, 10.1159/000089011

Montgomery, 2008, The search for genes contributing to endometriosis risk, Hum. Reprod. Update, 14, 447, 10.1093/humupd/dmn016

Ayaz, 2009, Role of CYP2C19 polymorphisms in patients with endometriosis, Gynecol. Endocrinol., 25, 530, 10.1080/09513590902972059

Bozdag, 2010, CYP17 and CYP2C19 gene polymorphisms in patients with endometriosis, Reprod. Biomed. Online, 20, 286, 10.1016/j.rbmo.2009.11.007

Guo, 2009, Reassessing the evidence for the link between dioxin and endometriosis: From molecular biology to clinical epidemiology, MHR Basic Sci. Reprod. Med., 15, 609, 10.1093/molehr/gap075

Ioannidis, 2001, Replication validity of genetic association studies, Nat. Genet., 29, 306, 10.1038/ng749

Lohmueller, 2003, Meta-analysis of genetic association studies supports a contribution of common variants to susceptibility to common disease, Nat. Genet., 33, 177, 10.1038/ng1071

Hindorff, 2009, Potential etiologic and functional implications of genome-wide association loci for human diseases and traits, Proc. Natl. Acad. Sci., 106, 9362, 10.1073/pnas.0903103106

McCarthy, 2008, Genome-wide association studies for complex traits: Consensus, uncertainty and challenges, Nat. Rev. Genet., 9, 356, 10.1038/nrg2344

Visscher, 2009, Genome-wide association studies and human disease: From trickle to flood, JAMA, 302, 2028, 10.1001/jama.2009.1643

Zondervan, 2002, The Genetic Epidemiology of Spontaneous Endometriosis in the Rhesus Monkey, Ann. N. Y. Acad. Sci., 955, 233, 10.1111/j.1749-6632.2002.tb02784.x

Zondervan, 2004, The complex interplay among factors that influence allelic association, Nat. Rev. Genet., 5, 89, 10.1038/nrg1270

Hirschhorn, 2002, A comprehensive review of genetic association studies, Genet. Med., 4, 45, 10.1097/00125817-200203000-00002

Kruglyak, 1995, Complete multipoint sib-pair analysis of qualitative and quantitative traits, Am. J. Hum. Genet., 57, 439

Lander, 1995, Genetic dissection of complex traits: Guidelines for interpreting and reporting linkage results, Nat. Genet., 11, 241, 10.1038/ng1195-241

Risch, 1990, Linkage strategies for genetically complex traits. II. The power of affected relative pairs, Am. J. Hum. Genet., 46, 229

Treloar, 2005, 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, 365, 10.1086/432960

Miki, 1994, A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1, Science, 266, 66, 10.1126/science.7545954

Wooster, 1995, Identification of the breast cancer susceptibility gene BRCA2, Nature, 378, 789, 10.1038/378789a0

Zondervan, 2007, Significant evidence of one or more susceptibility loci for endometriosis with near-Mendelian inheritance on chromosome 7p13–15, Hum. Reprod., 22, 717, 10.1093/humrep/del446

(2007). The International HapMap Consortium A second generation human haplotype map of over 3.1 million SNPs. Nature, 449, 851–861.

Treloar, 2007, Variants in EMX2 and PTEN do not contribute to risk of endometriosis, MHR Basic Sci. Reprod. Med., 13, 587, 10.1093/molehr/gam023

Daftary, 2004, EMX2 Gene Expression in the Female Reproductive Tract and Aberrant Expression in the Endometrium of Patients with Endometriosis, J. Clin. Endocrinol. Metab., 89, 2390, 10.1210/jc.2003-031389

Troy, 2003, Transcriptional Repression of Peri-Implantation EMX2 Expression in Mammalian Reproduction by HOXA10, Mol. Cell. Biol., 23, 1, 10.1128/MCB.23.1.1-13.2003

Maxwell, 1998, Mutation of the PTEN Tumor Suppressor Gene in Endometrial Hyperplasias, Cancer Res., 58, 2500

Easton, 2007, Genome-wide association study identifies novel breast cancer susceptibility loci, Nature, 447, 1087, 10.1038/nature05887

Pollock, 2007, Frequent activating FGFR2 mutations in endometrial carcinomas parallel germline mutations associated with craniosynostosis and skeletal dysplasia syndromes, Oncogene, 26, 7158, 10.1038/sj.onc.1210529

Zhao, 2008, Common variation in the fibroblast growth factor receptor 2 gene is not associated with endometriosis risk, Hum. Reprod., 23, 1661, 10.1093/humrep/den035

Zondervan, 2007, Designing candidate gene and genome-wide case–control association studies, Nat. Protoc., 2, 2492, 10.1038/nprot.2007.366

Pritchard, 2007, Overcoming the Winner’s Curse: Estimating Penetrance Parameters from Case-Control Data, Am. J. Hum. Genet., 80, 605, 10.1086/512821

Sabeti, 2007, Genome-wide detection and characterization of positive selection in human populations, Nature, 449, 913, 10.1038/nature06250

Uno, 2010, A genome-wide association study identifies genetic variants in the CDKN2BAS locus associated with endometriosis in Japanese, Nat. Genet., 42, 707, 10.1038/ng.612

Sikora, 2005, Concentrations of interleukin (IL)-1α, IL-1 soluble receptor type II (IL-1 sRII) and IL-1 receptor antagonist (IL-1 Ra) in the peritoneal fluid and serum of infertile women with endometriosis, Eur. J. Obstet. Gynecol. Reprod. Biol., 123, 198, 10.1016/j.ejogrb.2005.04.019

Adachi, 2010, Meta-analysis of genome-wide association scans for genetic susceptibility to endometriosis in Japanese population, J. Hum. Genet., 55, 816, 10.1038/jhg.2010.118

Zanatta, 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, 10.1007/s10815-010-9471-y

Naillat, 2010, Wnt4/5a signalling coordinates cell adhesion and entry into meiosis during presumptive ovarian follicle development, Hum. Mol. Genet., 19, 1539, 10.1093/hmg/ddq027

Boyer, 2010, WNT4 is required for normal ovarian follicle development and female fertility, FASEB J., 24, 3010, 10.1096/fj.09-145789

MacDonald, 2009, Wnt/β-Catenin Signaling: Components, Mechanisms, and Diseases, Dev. Cell, 17, 9, 10.1016/j.devcel.2009.06.016

Yang, 2010, Common SNPs explain a large proportion of the heritability for human height, Nat. Genet., 42, 565, 10.1038/ng.608

Painter, 2011, Genome-wide association study identifies a locus at 7p15.2 associated with endometriosis, Nat. Genet., 43, 51, 10.1038/ng.731

Bodmer, 2008, Common and rare variants in multifactorial susceptibility to common diseases, Nat. Genet., 40, 695, 10.1038/ng.f.136

Gorlov, 2008, Shifting Paradigm of Association Studies: Value of Rare Single-Nucleotide Polymorphisms, Am. J. Hum. Genet., 82, 100, 10.1016/j.ajhg.2007.09.006

Curtin, 2009, Identifying Rarer Genetic Variants for Common Complex Diseases: Diseased Versus Neutral Discovery Panels, Ann. Hum. Genet., 73, 54, 10.1111/j.1469-1809.2008.00483.x

Iles, M.M. (2008). What Can Genome-Wide Association Studies Tell Us about the Genetics of Common Disease?. PLOS Genet., 4.

Mettler, 2007, Comparison of c-DNA microarray analysis of gene expression between eutopic endometrium and ectopic endometrium (endometriosis), J. Assist. Reprod. Genet., 24, 249, 10.1007/s10815-007-9116-y

Eyster, 2007, Whole genome deoxyribonucleic acid microarray analysis of gene expression in ectopic versus eutopic endometrium, Fertil. Steril., 88, 1505, 10.1016/j.fertnstert.2007.01.056

Flores, 2007, Molecular profiling of experimental endometriosis identified gene expression patterns in common with human disease, Fertil. Steril., 87, 1180, 10.1016/j.fertnstert.2006.07.1550

Hull, 2008, Endometrial-Peritoneal Interactions during Endometriotic Lesion Establishment, Am. J. Pathol., 173, 700, 10.2353/ajpath.2008.071128

Zafrakas, 2008, Genome-wide microarray gene expression, array-CGH analysis, and telomerase activity in advanced ovarian endometriosis: A high degree of differentiation rather than malignant potential, Int. J. Mol. Med., 21, 335

Pelch, 2010, Aberrant gene expression profile in a mouse model of endometriosis mirrors that observed in women, Fertil. Steril., 93, 1615, 10.1016/j.fertnstert.2009.03.086

Umezawa, 2009, Microarray analysis provides insight into the early steps of pathophysiology of mouse endometriosis model induced by autotransplantation of endometrium, Life Sci., 84, 832, 10.1016/j.lfs.2009.03.015

Gnecco, 2017, Exposure to the environmental endocrine disruptor TCDD and human reproductive dysfunction: Translating lessons from murine models, Reprod. Toxicol. Elmsford N, 68, 59, 10.1016/j.reprotox.2016.07.007

Morgan, 2005, Epigenetic reprogramming in mammals, Hum. Mol. Genet., 14, R47, 10.1093/hmg/ddi114

Gabory, 2011, Epigenetic mechanisms involved in developmental nutritional programming, World J. Diabetes, 2, 164, 10.4239/wjd.v2.i10.164

Berger, 2009, An operational definition of epigenetics, Genes Dev., 23, 781, 10.1101/gad.1787609

Robertson, 2005, DNA methylation and human disease, Nat. Rev. Genet., 6, 597, 10.1038/nrg1655

Robertson, 2000, DNA methylation in health and disease, Nat. Rev. Genet., 1, 11, 10.1038/35049533

Rodenhiser, 2006, Epigenetics and human disease: Translating basic biology into clinical applications, CMAJ Can. Med. Assoc. J. J. Assoc. Medicale Can., 174, 341, 10.1503/cmaj.050774

Li, 2002, Chromatin modification and epigenetic reprogramming in mammalian development, Nat. Rev. Genet., 3, 662, 10.1038/nrg887

Ritchie, 2017, microRNA Target Prediction, Methods Mol. Biol. Clifton NJ, 1513, 193, 10.1007/978-1-4939-6539-7_13

Sun, 2013, Adult-specific functions of animal microRNAs, Nat. Rev. Genet., 14, 535, 10.1038/nrg3471

Kurokawa, 2009, Transcriptional regulation through noncoding RNAs and epigenetic modifications, RNA Biol., 6, 233, 10.4161/rna.6.3.8329

Saare, 2017, Challenges in endometriosis miRNA studies - From tissue heterogeneity to disease specific miRNAs, Biochim. Biophys. Acta Mol. Basis Dis., 1863, 2282, 10.1016/j.bbadis.2017.06.018

Munro, 2010, Epigenetic regulation of endometrium during the menstrual cycle, Mol. Hum. Reprod., 16, 297, 10.1093/molehr/gaq010

Yamagata, 2009, DNA methyltransferase expression in the human endometrium: Down-regulation by progesterone and estrogen, Hum. Reprod. Oxf. Engl., 24, 1126, 10.1093/humrep/dep015

Krusche, 2007, Class I histone deacetylase expression in the human cyclic endometrium and endometrial adenocarcinomas, Hum. Reprod. Oxf. Engl., 22, 2956, 10.1093/humrep/dem241

Sakai, 2003, Involvement of histone acetylation in ovarian steroid-induced decidualization of human endometrial stromal cells, J. Biol. Chem., 278, 16675, 10.1074/jbc.M211715200

Uchida, 2007, Histone deacetylase inhibitor-induced glycodelin enhances the initial step of implantation, Hum. Reprod. Oxf. Engl., 22, 2615, 10.1093/humrep/dem263

Uchida, 2006, Human endometrial cytodifferentiation by histone deacetylase inhibitors, Hum. Cell, 19, 38, 10.1111/j.1749-0774.2005.00006.x

Galliano, 2014, MicroRNA and implantation, Fertil. Steril., 101, 1531, 10.1016/j.fertnstert.2014.04.023

Hull, 2013, Tissue and circulating microRNA influence reproductive function in endometrial disease, Reprod. Biomed. Online, 27, 515, 10.1016/j.rbmo.2013.07.012

Pan, 2008, MicroRNA signature and regulatory functions in the endometrium during normal and disease states, Semin. Reprod. Med., 26, 479, 10.1055/s-0028-1096128

Kuokkanen, 2010, Genomic profiling of microRNAs and messenger RNAs reveals hormonal regulation in microRNA expression in human endometrium, Biol. Reprod., 82, 791, 10.1095/biolreprod.109.081059

Lessey, 2010, Fine tuning of endometrial function by estrogen and progesterone through microRNAs, Biol. Reprod., 82, 653, 10.1095/biolreprod.110.083667

Pastorelli, 2009, Genetic analyses reveal a requirement for Dicer1 in the mouse urogenital tract, Mamm. Genome Off. J. Int. Mamm. Genome Soc., 20, 140, 10.1007/s00335-008-9169-y

Hong, 2008, Dicer1 is essential for female fertility and normal development of the female reproductive system, Endocrinology, 149, 6207, 10.1210/en.2008-0294

Koninckx, 2019, Pathogenesis of endometriosis: The genetic/epigenetic theory, Fertil. Steril., 111, 327, 10.1016/j.fertnstert.2018.10.013

Yamagata, Y., Nishino, K., Takaki, E., Sato, S., Maekawa, R., Nakai, A., and Sugino, N. (2014). Genome-wide DNA methylation profiling in cultured eutopic and ectopic endometrial stromal cells. PloS ONE, 9.

Signorile, P.G., Severino, A., Santoro, M., Spyrou, M., Viceconte, R., and Baldi, A. (2018). Methylation analysis of HOXA10 regulatory elements in patients with endometriosis. BMC Res. Notes, 11.

Wu, 2005, Aberrant methylation at HOXA10 may be responsible for its aberrant expression in the endometrium of patients with endometriosis, Am. J. Obstet. Gynecol., 193, 371, 10.1016/j.ajog.2005.01.034

Lee, 2009, Experimental Murine Endometriosis Induces DNA Methylation and Altered Gene Expression in Eutopic Endometrium, Biol. Reprod., 80, 79, 10.1095/biolreprod.108.070391

Bromer, 2009, Hypermethylation of Homeobox A10 by in Utero Diethylstilbestrol Exposure: An Epigenetic Mechanism for Altered Developmental Programming, Endocrinology, 150, 3376, 10.1210/en.2009-0071

Bromer, 2010, Bisphenol-A exposure in utero leads to epigenetic alterations in the developmental programming of uterine estrogen response, FASEB J., 24, 2273, 10.1096/fj.09-140533

Kulp, 2016, Aberrant HOXA10 Methylation in Patients With Common Gynecologic Disorders, Reprod. Sci., 23, 455, 10.1177/1933719116630427

Wu, 2006, Promoter hypermethylation of progesterone receptor isoform B (PR-B) in endometriosis, Epigenetics, 1, 106, 10.4161/epi.1.2.2766

Attia, 2000, Progesterone receptor isoform A but not B is expressed in endometriosis, J. Clin. Endocrinol. Metab., 85, 2897

Nie, 2010, Promoter hypermethylation of progesterone receptor isoform B (PR-B) in adenomyosis and its rectification by a histone deacetylase inhibitor and a demethylation agent, Reprod. Sci. Thousand Oaks Calif, 17, 995, 10.1177/1933719110377118

Wu, 2007, Aberrant expression of deoxyribonucleic acid methyltransferases DNMT1, DNMT3A, and DNMT3B in women with endometriosis, Fertil. Steril., 87, 24, 10.1016/j.fertnstert.2006.05.077

Dyson, 2015, Aberrant expression and localization of deoxyribonucleic acid methyltransferase 3B in endometriotic stromal cells, Fertil. Steril., 104, 953, 10.1016/j.fertnstert.2015.06.046

Aznaurova, 2014, Molecular aspects of development and regulation of endometriosis, Reprod. Biol. Endocrinol. RBE, 12, 50, 10.1186/1477-7827-12-50

Wu, 2007, Trichostatin A, a histone deacetylase inhibitor, attenuates invasiveness and reactivates E-cadherin expression in immortalized endometriotic cells, Reprod. Sci. Thousand Oaks Calif, 14, 374, 10.1177/1933719107302913

KOUKOURA, 2016, DNA methylation in endometriosis (Review), Mol. Med. Rep., 13, 2939, 10.3892/mmr.2016.4925

Arosh, 2015, Selective inhibition of prostaglandin E2 receptors EP2 and EP4 modulates DNA methylation and histone modification machinery proteins in human endometriotic cells, Mol. Cell. Endocrinol., 409, 51, 10.1016/j.mce.2015.03.023

Xue, 2007, Transcriptional activation of steroidogenic factor-1 by hypomethylation of the 5’ CpG island in endometriosis, J. Clin. Endocrinol. Metab., 92, 3261, 10.1210/jc.2007-0494

Xue, 2007, Promoter methylation regulates estrogen receptor 2 in human endometrium and endometriosis, Biol. Reprod., 77, 681, 10.1095/biolreprod.107.061804

Izawa, 2011, Demethylation of a nonpromoter cytosine-phosphate-guanine island in the aromatase gene may cause the aberrant up-regulation in endometriotic tissues, Fertil. Steril., 95, 33, 10.1016/j.fertnstert.2010.06.024

Wang, 2012, DNA hypomethylation of the COX-2 gene promoter is associated with up-regulation of its mRNA expression in eutopic endometrium of endometriosis, Eur. J. Med. Res., 17, 12, 10.1186/2047-783X-17-12

Nasu, 2014, Aberrant histone modification in endometriosis, Front. Biosci. Landmark Ed., 19, 1202, 10.2741/4276

Monteiro, 2014, Endometriosis Is Characterized by a Distinct Pattern of Histone 3 and Histone 4 Lysine Modifications, Reprod. Sci., 21, 305, 10.1177/1933719113497267

Xiaomeng, 2013, Aberrant histone acetylation and methylation levels in woman with endometriosis, Arch. Gynecol. Obstet., 287, 487, 10.1007/s00404-012-2591-0

Samartzis, 2013, The Expression of Histone Deacetylase 1, But Not Other Class I Histone Deacetylases, Is Significantly Increased in Endometriosis, Reprod. Sci., 20, 1416, 10.1177/1933719113488450

Kawano, 2011, Application of the histone deacetylase inhibitors for the treatment of endometriosis: Histone modifications as pathogenesis and novel therapeutic target, Hum. Reprod. Oxf. Engl., 26, 2486, 10.1093/humrep/der203

Imesch, 2011, Inhibition of transcription, expression, and secretion of the vascular epithelial growth factor in human epithelial endometriotic cells by romidepsin, Fertil. Steril., 95, 1579, 10.1016/j.fertnstert.2010.12.058

Ayhan, 2012, Loss of ARID1A expression is an early molecular event in tumor progression from ovarian endometriotic cyst to clear cell and endometrioid carcinoma, Int. J. Gynecol. Cancer Off. J. Int. Gynecol. Cancer Soc., 22, 1310, 10.1097/IGC.0b013e31826b5dcc

Lowery, 2012, Loss of ARID1A-associated Protein Expression Is a Frequent Event in Clear Cell and Endometrioid Ovarian Cancers, Int. J. Gynecol. Cancer, 22, 9, 10.1097/IGC.0b013e318231f140

Chene, 2015, The ARID1A pathway in ovarian clear cell and endometrioid carcinoma, contiguous endometriosis, and benign endometriosis, Int. J. Gynaecol. Obstet. Off. Organ Int. Fed. Gynaecol. Obstet., 130, 27, 10.1016/j.ijgo.2015.02.021

Er, 2016, Targeted next-generation sequencing for molecular diagnosis of endometriosis-associated ovarian cancer, J. Mol. Med. Berl. Ger., 94, 835, 10.1007/s00109-016-1395-2

Nothnick, 2017, MicroRNAs and Endometriosis: Distinguishing Drivers from Passengers in Disease Pathogenesis, Semin. Reprod. Med., 35, 173, 10.1055/s-0037-1599089

Filigheddu, N., Gregnanin, I., Porporato, P.E., Surico, D., Perego, B., Galli, L., Patrignani, C., Graziani, A., and Surico, N. (2010). Differential Expression of MicroRNAs between Eutopic and Ectopic Endometrium in Ovarian Endometriosis. J. Biomed. Biotechnol., 2010.

Burney, 2009, MicroRNA expression profiling of eutopic secretory endometrium in women with versus without endometriosis, Mol. Hum. Reprod., 15, 625, 10.1093/molehr/gap068

Vasudevan, 2007, Switching from repression to activation: MicroRNAs can up-regulate translation, Science, 318, 1931, 10.1126/science.1149460

Han, 2017, Risk factors and biomarkers for the recurrence of ovarian endometrioma: About the immunoreactivity of progesterone receptor isoform B and nuclear factor kappa B, Gynecol. Endocrinol. Off. J. Int. Soc. Gynecol. Endocrinol., 33, 70, 10.1080/09513590.2016.1205580

Wu, 2006, Inhibition of Proliferation of Endometrial Stromal Cells by Trichostatin A, RU486, CDB-2914, N-Acetylcysteine, and ICI 182780, Gynecol. Obstet. Investig., 62, 193, 10.1159/000093975

Wu, 2008, Histone deacetylase inhibitors trichostatin A and valproic acid induce cell cycle arrest and p21 expression in immortalized human endometrial stromal cells, Eur. J. Obstet. Gynecol. Reprod. Biol., 137, 198, 10.1016/j.ejogrb.2007.02.014

Wu, 2007, Suppression of IL-1β-induced COX-2 expression by trichostatin A (TSA) in human endometrial stromal cells, Eur. J. Obstet. Gynecol. Reprod. Biol., 135, 88, 10.1016/j.ejogrb.2006.07.034

Wu, 2009, Peroxisome proliferator-activated receptor-gamma and retinoid X receptor agonists synergistically suppress proliferation of immortalized endometrial stromal cells, Fertil. Steril., 91, 2142, 10.1016/j.fertnstert.2008.04.012

Peeters, 2006, PPARgamma represses VEGF expression in human endometrial cells: Implications for uterine angiogenesis, Angiogenesis, 8, 373, 10.1007/s10456-005-9027-4

Ohama, 2008, Peroxisome proliferator-activated receptor-γ ligand reduced tumor necrosis factor-α-induced interleukin-8 production and growth in endometriotic stromal cells, Fertil. Steril., 89, 311, 10.1016/j.fertnstert.2007.03.061

Lu, 2010, Trichostatin A, a histone deacetylase inhibitor, reduces lesion growth and hyperalgesia in experimentally induced endometriosis in mice, Hum. Reprod. Oxf. Engl., 25, 1014, 10.1093/humrep/dep472

Imesch, 2010, Romidepsin reduces histone deacetylase activity, induces acetylation of histones, inhibits proliferation, and activates apoptosis in immortalized epithelial endometriotic cells, Fertil. Steril., 94, 2838, 10.1016/j.fertnstert.2010.04.052

Wang, 2007, Histone Deacetylase Inhibitors Suppress TF-κB-dependent Agonist-driven Tissue Factor Expression in Endothelial Cells and Monocytes, J. Biol. Chem., 282, 28408, 10.1074/jbc.M703586200

Dong, 2007, Histone deacetylase inhibitor valproic acid inhibits proliferation and induces apoptosis in KM3 cells via downregulating VEGF receptor, Neuro Endocrinol. Lett., 28, 775

Deroanne, 2002, Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling, Oncogene, 21, 427, 10.1038/sj.onc.1205108

Lockwood, 2009, Decidualized Human Endometrial Stromal Cells Mediate Hemostasis, Angiogenesis, and Abnormal Uterine Bleeding, Reprod. Sci. Thousand Oaks Calif, 16, 162, 10.1177/1933719108325758

Liu, 2008, A pilot study on the off-label use of valproic acid to treat adenomyosis, Fertil. Steril., 89, 246, 10.1016/j.fertnstert.2006.11.009

Liu, 2010, Valproic acid as a therapy for adenomyosis: A comparative case series, Reprod. Sci. Thousand Oaks Calif, 17, 904, 10.1177/1933719110373807

Liu, 2012, Valproic acid and progestin inhibit lesion growth and reduce hyperalgesia in experimentally induced endometriosis in rats, Reprod. Sci. Thousand Oaks Calif, 19, 360, 10.1177/1933719111424453

Barra, F., and Ferrero, S. (2018). Epigenetic Drugs in the Treatment of Endometriosis. Reprod. Sci. Thousand Oaks Calif, 1933719118765987.