Review Article

多囊卵巢综合征的表观遗传标记

卷 27, 期 39, 2020

页: [6727 - 6743] 页: 17

弟呕挨: 10.2174/0929867326666191003154548

价格: $65

conference banner
摘要

多囊卵巢综合症(PCOS)是一种常见的内分泌和代谢疾病,会影响育龄妇女。 最近的研究表明,基因在PCOS的病因中具有重要作用。 但是,人们对这些基因转录和转录后调控的确切方式知之甚少。 本审查的目的是提供有关miRNA和DNA甲基化作为PCOS的表观遗传标记的最新信息。 此处提供的数据可以得出结论,在选择特定PCOS表型的治疗方法时,可以将microRNA和DNA甲基化视为可能有用的生物标记,因此代表了诊断和治疗PCOS患者的两个重要工具。

关键词: 多囊卵巢综合征,表观遗传标记,miRNA,DNA甲基化,PCOS表型,PCOS治疗。

[1]
Azziz, R.; Carmina, E.; Dewailly, D.; Diamanti-Kandarakis, E.; Escobar-Morreale, H.F.; Futterweit, W.; Janssen, O.E.; Legro, R.S.; Norman, R.J.; Taylor, A.E.; Witchel, S.F. Task force on the phenotype of the polycystic ovary syndrome of the androgen excess and PCOS society. The androgen excess and PCOS Society criteria for the polycystic ovary syndrome: the complete task force report. Fertil. Steril., 2009, 91(2), 456-488.
[http://dx.doi.org/10.1016/j.fertnstert.2008.06.035] [PMID: 18950759]
[2]
Rotterdam ESHRE/ASRM-sponsored PCOS consensus workshop group. Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome. Fertil. Steril., 2004, 81(1), 19-25.
[http://dx.doi.org/10.1016/j.fertnstert.2003.10.004] [PMID: 14711538]
[3]
Zawadzki, J.K.; Dunaif, A. Diagnostic criteria of polycystic ovary syndrome: towards a rational approach in: Polycystic Ovary Syndrome; Dunaif A.; Givens, J.R.; Haseline, F.P; Merriam, G.R., Ed.; Blackwell Scientific Publications: Boston, MA, USA, 1992.
[4]
Franks, S.; Stark, J.; Hardy, K. Follicle dynamics and anovulation in polycystic ovary syndrome. Hum. Reprod. Update, 2008, 14(4), 367-378.
[http://dx.doi.org/10.1093/humupd/dmn015] [PMID: 18499708]
[5]
Ferreira, S.R.; Motta, A.B. Uterine function: from normal to polycystic ovarian syndrome alterations. Curr. Med. Chem., 2018, 25(15), 1792-1804.
[http://dx.doi.org/10.2174/0929867325666171205144119] [PMID: 29210631]
[6]
Franks, S. Polycystic ovary syndrome. N. Engl. J. Med., 1995, 333(13), 853-861.
[http://dx.doi.org/10.1056/NEJM199509283331307] [PMID: 7651477]
[7]
Dunaif, A. Insulin resistance and the polycystic ovary syndrome: mechanism and implications for pathogenesis. Endocr. Rev., 1997, 18(6), 774-800.
[http://dx.doi.org/10.1210/edrv.18.6.0318 ] [PMID: 9408743]
[8]
Subramaniam, K.; Tripathi, A.; Dabadghao, P. Familial clustering of metabolic phenotype in brothers of women with polycystic ovary syndrome. Gynecol. Endocrinol., 2019, 35(7), 601-603.
[http://dx.doi.org/10.1080/09513590.2019.1566451] [PMID: 30727783]
[9]
Yilmaz, B.; Vellanki, P.; Ata, B.; Yildiz, B.O. Diabetes mellitus and insulin resistance in mothers, fathers, sisters, and brothers of women with polycystic ovary syndrome: a systematic review and meta-analysis. Fertil. Steril., 2018, 110(3), 523-533.e14.
[http://dx.doi.org/10.1016/j.fertnstert.2018.04.024] [PMID: 29960703]
[10]
Torchen, L.C.; Kumar, A.; Kalra, B.; Savjani, G.; Sisk, R.; Legro, R.S.; Dunaif, A. Increased antimüllerian hormone levels and other reproductive endocrine changes in adult male relatives of women with polycystic ovary syndrome. Fertil. Steril., 2016, 106(1), 50-55.
[http://dx.doi.org/10.1016/j.fertnstert.2016.03.029] [PMID: 27042970]
[11]
Chen, Z-J.; Zhao, H.; He, L.; Shi, Y.; Qin, Y.; Shi, Y.; Li, Z.; You, L.; Zhao, J.; Liu, J.; Liang, X.; Zhao, X.; Zhao, J.; Sun, Y.; Zhang, B.; Jiang, H.; Zhao, D.; Bian, Y.; Gao, X.; Geng, L.; Li, Y.; Zhu, D.; Sun, X.; Xu, J.E.; Hao, C.; Ren, C.E.; Zhang, Y.; Chen, S.; Zhang, W.; Yang, A.; Yan, J.; Li, Y.; Ma, J.; Zhao, Y. Genome-wide association study identifies susceptibility loci for polycystic ovary syndrome on chromosome 2p16.3, 2p21 and 9q33.3. Nat. Genet., 2011, 43(1), 55-59.
[http://dx.doi.org/10.1038/ng.732] [PMID: 21151128]
[12]
Shi, Y.; Zhao, H.; Shi, Y.; Cao, Y.; Yang, D.; Li, Z.; Zhang, B.; Liang, X.; Li, T.; Chen, J.; Shen, J.; Zhao, J.; You, L.; Gao, X.; Zhu, D.; Zhao, X.; Yan, Y.; Qin, Y.; Li, W.; Yan, J.; Wang, Q.; Zhao, J.; Geng, L.; Ma, J.; Zhao, Y.; He, G.; Zhang, A.; Zou, S.; Yang, A.; Liu, J.; Li, W.; Li, B.; Wan, C.; Qin, Y.; Shi, J.; Yang, J.; Jiang, H.; Xu, J.E.; Qi, X.; Sun, Y.; Zhang, Y.; Hao, C.; Ju, X.; Zhao, D.; Ren, C.E.; Li, X.; Zhang, W.; Zhang, Y.; Zhang, J.; Wu, D.; Zhang, C.; He, L.; Chen, Z.J. Genome-wide association study identifies eight new risk loci for polycystic ovary syndrome. Nat. Genet., 2012, 44(9), 1020-1025.
[http://dx.doi.org/10.1038/ng.2384] [PMID: 22885925]
[13]
Toledo, S.P.; Brunner, H.G.; Kraaij, R.; Post, M.; Dahia, P.L.; Hayashida, C.Y.; Kremer, H.; Themmen, A.P.; Themmen, A.P. An inactivating mutation of the luteinizing hormone receptor causes amenorrhea in a 46,XX female. J. Clin. Endocrinol. Metab., 1996, 81(11), 3850-3854.
[http://dx.doi.org/10.1210/jcem.81.11.8923827 ] [PMID: 8923827]
[14]
Latronico, A.C.; Lins, T.S.; Brito, V.N.; Arnhold, I.J.; Mendonca, B.B. The effect of distinct activating mutations of the luteinizing hormone receptor gene on the pituitary-gonadal axis in both sexes. Clin. Endocrinol. (Oxf.), 2000, 53(5), 609-613.
[http://dx.doi.org/10.1046/j.1365-2265.2000.01135.x] [PMID: 11106922]
[15]
McAllister, J.M.; Legro, R.S.; Modi, B.P.; Strauss, J.F. III Functional genomics of PCOS: from GWAS to molecular mechanisms. Trends Endocrinol. Metab., 2015, 26(3), 118-124.
[http://dx.doi.org/10.1016/j.tem.2014.12.004] [PMID: 25600292]
[16]
Mutharasan, P.; Galdones, E.; Peñalver Bernabé, B.; Garcia, O.A.; Jafari, N.; Shea, L.D.; Woodruff, T.K.; Legro, R.S.; Dunaif, A.; Urbanek, M. Evidence for chromosome 2p16.3 polycystic ovary syndrome susceptibility locus in affected women of European ancestry. J. Clin. Endocrinol. Metab., 2013, 98(1), E185-E190.
[http://dx.doi.org/10.1210/jc.2012-2471] [PMID: 23118426]
[17]
Moller, D.E.; Yokota, A.; White, M.F.; Pazianos, A.G.; Flier, J.S. A naturally occurring mutation of insulin receptor alanine 1134 impairs tyrosine kinase function and is associated with dominantly inherited insulin resistance. J. Biol. Chem., 1990, 265(25), 14979-14985.
[PMID: 2168397]
[18]
Saxena, R.; Elbers, C.C.; Guo, Y.; Peter, I.; Gaunt, T.R.; Mega, J.L.; Lanktree, M.B.; Tare, A.; Castillo, B.A.; Li, Y.R.; Johnson, T.; Bruinenberg, M.; Gilbert-Diamond, D.; Rajagopalan, R.; Voight, B.F.; Balasubramanyam, A.; Barnard, J.; Bauer, F.; Baumert, J.; Bhangale, T.; Böhm, B.O.; Braund, P.S.; Burton, P.R.; Chandrupatla, H.R.; Clarke, R.; Cooper-DeHoff, R.M.; Crook, E.D.; Davey-Smith, G.; Day, I.N.; de Boer, A.; de Groot, M.C.; Drenos, F.; Ferguson, J.; Fox, C.S.; Furlong, C.E.; Gibson, Q.; Gieger, C.; Gilhuijs-Pederson, L.A.; Glessner, J.T.; Goel, A.; Gong, Y.; Grant, S.F.; Grobbee, D.E.; Hastie, C.; Humphries, S.E.; Kim, C.E.; Kivimaki, M.; Kleber, M.; Meisinger, C.; Kumari, M.; Langaee, T.Y.; Lawlor, D.A.; Li, M.; Lobmeyer, M.T.; Maitland-van der Zee, A.H.; Meijs, M.F.; Molony, C.M.; Morrow, D.A.; Murugesan, G.; Musani, S.K.; Nelson, C.P.; Newhouse, S.J.; O’Connell, J.R.; Padmanabhan, S.; Palmen, J.; Patel, S.R.; Pepine, C.J.; Pettinger, M.; Price, T.S.; Rafelt, S.; Ranchalis, J.; Rasheed, A.; Rosenthal, E.; Ruczinski, I.; Shah, S.; Shen, H.; Silbernagel, G.; Smith, E.N.; Spijkerman, A.W.; Stanton, A.; Steffes, M.W.; Thorand, B.; Trip, M.; van der Harst, P. van der A, D.L.; van Iperen, E.P.; van Setten, J.; van Vliet-Ostaptchouk, J.V.; Verweij, N.; Wolffenbuttel, B.H.; Young, T.; Zafarmand, M.H.; Zmuda, J.M.; Boehnke, M.; Altshuler, D.; McCarthy, M.; Kao, W.H.; Pankow, J.S.; Cappola, T.P.; Sever, P.; Poulter, N.; Caulfield, M.; Dominiczak, A.; Shields, D.C.; Bhatt, D.L.; Zhang, L.; Curtis, S.P.; Danesh, J.; Casas, J.P.; van der Schouw, Y.T.; Onland-Moret, N.C.; Doevendans, P.A.; Dorn, G.W., II; Farrall, M.; FitzGerald, G.A.; Hamsten, A.; Hegele, R.; Hingorani, A.D.; Hofker, M.H.; Huggins, G.S.; Illig, T.; Jarvik, G.P.; Johnson, J.A.; Klungel, O.H.; Knowler, W.C.; Koenig, W.; März, W.; Meigs, J.B.; Melander, O.; Munroe, P.B.; Mitchell, B.D.; Bielinski, S.J.; Rader, D.J.; Reilly, M.P.; Rich, S.S.; Rotter, J.I.; Saleheen, D.; Samani, N.J.; Schadt, E.E.; Shuldiner, A.R.; Silverstein, R.; Kottke-Marchant, K.; Talmud, P.J.; Watkins, H.; Asselbergs, F.W.; de Bakker, P.I.; McCaffery, J.; Wijmenga, C.; Sabatine, M.S.; Wilson, J.G.; Reiner, A.; Bowden, D.W.; Hakonarson, H.; Siscovick, D.S.; Keating, B.J. Large-scale gene-centric meta-analysis across 39 studies identifies type 2 diabetes loci. Am. J. Hum. Genet., 2012, 90(3), 410-425.
[http://dx.doi.org/10.1016/j.ajhg.2011.12.022] [PMID: 22325160]
[19]
Voight, B.F.; Scott, L.J.; Steinthorsdottir, V.; Morris, A.P.; Dina, C.; Welch, R.P.; Zeggini, E.; Huth, C.; Aulchenko, Y.S.; Thorleifsson, G.; McCulloch, L.J.; Ferreira, T.; Grallert, H.; Amin, N.; Wu, G.; Willer, C.J.; Raychaudhuri, S.; McCarroll, S.A.; Langenberg, C.; Hofmann, O.M.; Dupuis, J.; Qi, L.; Segrè, A.V.; van Hoek, M.; Navarro, P.; Ardlie, K.; Balkau, B.; Benediktsson, R.; Bennett, A.J.; Blagieva, R.; Boerwinkle, E.; Bonnycastle, L.L.; Bengtsson Boström, K.; Bravenboer, B.; Bumpstead, S.; Burtt, N.P.; Charpentier, G.; Chines, P.S.; Cornelis, M.; Couper, D.J.; Crawford, G.; Doney, A.S.; Elliott, K.S.; Elliott, A.L.; Erdos, M.R.; Fox, C.S.; Franklin, C.S.; Ganser, M.; Gieger, C.; Grarup, N.; Green, T.; Griffin, S.; Groves, C.J.; Guiducci, C.; Hadjadj, S.; Hassanali, N.; Herder, C.; Isomaa, B.; Jackson, A.U.; Johnson, P.R.; Jørgensen, T.; Kao, W.H.; Klopp, N.; Kong, A.; Kraft, P.; Kuusisto, J.; Lauritzen, T.; Li, M.; Lieverse, A.; Lindgren, C.M.; Lyssenko, V.; Marre, M.; Meitinger, T.; Midthjell, K.; Morken, M.A.; Narisu, N.; Nilsson, P.; Owen, K.R.; Payne, F.; Perry, J.R.; Petersen, A.K.; Platou, C.; Proença, C.; Prokopenko, I.; Rathmann, W.; Rayner, N.W.; Robertson, N.R.; Rocheleau, G.; Roden, M.; Sampson, M.J.; Saxena, R.; Shields, B.M.; Shrader, P.; Sigurdsson, G.; Sparsø, T.; Strassburger, K.; Stringham, H.M.; Sun, Q.; Swift, A.J.; Thorand, B.; Tichet, J.; Tuomi, T.; van Dam, R.M.; van Haeften, T.W.; van Herpt, T.; van Vliet-Ostaptchouk, J.V.; Walters, G.B.; Weedon, M.N.; Wijmenga, C.; Witteman, J.; Bergman, R.N.; Cauchi, S.; Collins, F.S.; Gloyn, A.L.; Gyllensten, U.; Hansen, T.; Hide, W.A.; Hitman, G.A.; Hofman, A.; Hunter, D.J.; Hveem, K.; Laakso, M.; Mohlke, K.L.; Morris, A.D.; Palmer, C.N.; Pramstaller, P.P.; Rudan, I.; Sijbrands, E.; Stein, L.D.; Tuomilehto, J.; Uitterlinden, A.; Walker, M.; Wareham, N.J.; Watanabe, R.M.; Abecasis, G.R.; Boehm, B.O.; Campbell, H.; Daly, M.J.; Hattersley, A.T.; Hu, F.B.; Meigs, J.B.; Pankow, J.S.; Pedersen, O.; Wichmann, H.E.; Barroso, I.; Florez, J.C.; Frayling, T.M.; Groop, L.; Sladek, R.; Thorsteinsdottir, U.; Wilson, J.F.; Illig, T.; Froguel, P.; van Duijn, C.M.; Stefansson, K.; Altshuler, D.; Boehnke, M.; McCarthy, M.I. MAGIC investigators. GIANT Consortium. Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis. Nat. Genet., 2010, 42(7), 579-589.
[http://dx.doi.org/10.1038/ng.609] [PMID: 20581827]
[20]
Rosenfield, R.L.; Ehrmann, D.A. The pathogenesis of polycystic ovary syndrome (PCOS): the hypothesis of PCOS as functional ovarian hyperandrogenism revisited. Endocr. Rev., 2016, 37(5), 467-520.
[http://dx.doi.org/10.1210/er.2015-1104] [PMID: 27459230]
[21]
McAllister, J.M.; Modi, B.; Miller, B.A.; Biegler, J.; Bruggeman, R.; Legro, R.S.; Strauss, J.F. III Overexpression of a DENND1A isoform produces a polycystic ovary syndrome theca phenotype. Proc. Natl. Acad. Sci. USA, 2014, 111(15), E1519-E1527.
[http://dx.doi.org/10.1073/pnas.1400574111] [PMID: 24706793]
[22]
Hayes, M.G.; Urbanek, M.; Ehrmann, D.A.; Armstrong, L.L.; Lee, J.Y.; Sisk, R.; Karaderi, T.; Barber, T.M.; McCarthy, M.I.; Franks, S.; Lindgren, C.M.; Welt, C.K.; Diamanti-Kandarakis, E.; Panidis, D.; Goodarzi, M.O.; Azziz, R.; Zhang, Y.; James, R.G.; Olivier, M.; Kissebah, A.H.; Stener-Victorin, E.; Legro, R.S.; Dunaif, A. Reproductive medicine network. Genome-wide association of polycystic ovary syndrome implicates alterations in gonadotropin secretion in European ancestry populations. Nat. Commun., 2015, 6, 7502.
[http://dx.doi.org/10.1038/ncomms8502] [PMID: 26284813]
[23]
Ren, B.; Guo, W.; Tang, Y.; Zhang, J.; Xiao, N.; Zhang, L.; Li, W. Rhein inhibits the migration of ovarian cancer cells through down-regulation of matrix metalloproteinases. Biol. Pharm. Bull., 2019, 42(4), 568-572.
[http://dx.doi.org/10.1248/bpb.b18-00431] [PMID: 30930417]
[24]
Peng, Y.; Zhang, W.; Yang, P.; Tian, Y.; Su, S.; Zhang, C.; Chen, Z.J.; Zhao, H. ERBB4 confers risk for polycystic ovary syndrome in han chinese. Sci. Rep., 2017, 7, 42000.
[http://dx.doi.org/10.1038/srep42000] [PMID: 28195137]
[25]
Crespo, R.P.; Bachega, T.A.S.S.; Mendonça, B.B.; Gomes, L.G. An update of genetic basis of PCOS pathogenesis. Arch. Endocrinol. Metab., 2018, 62(3), 352-361.
[http://dx.doi.org/10.20945/2359-3997000000049] [PMID: 29972435]
[26]
Jones, M.R.; Goodarzi, M.O. Genetic determinants of polycystic ovary syndrome: progress and future directions. Fertil. Steril., 2016, 106(1), 25-32.
[http://dx.doi.org/10.1016/j.fertnstert.2016.04.040] [PMID: 27179787]
[27]
de Bruin, C.; Dauber, A. Insights from exome sequencing for endocrine disorders. Nat. Rev. Endocrinol., 2015, 11(8), 455-464.
[http://dx.doi.org/10.1038/nrendo.2015.72] [PMID: 25963271]
[28]
Gorsic, L.K.; Kosova, G.; Werstein, B.; Sisk, R.; Legro, R.S.; Hayes, M.G.; Teixeira, J.M.; Dunaif, A.; Urbanek, M. pathogenic anti-müllerian hormone variants in polycystic ovary syndrome. J. Clin. Endocrinol. Metab., 2017, 102(8), 2862-2872.
[http://dx.doi.org/10.1210/jc.2017-00612] [PMID: 28505284]
[29]
Abbott, D.H.; Levine, J.E.; Dumesic, D.A. Translational insight into polycystic ovary syndrome (PCOS) from female monkeys with PCOS-like traits. Curr. Pharm. Des., 2016, 22(36), 5625-5633.
[http://dx.doi.org/10.2174/1381612822666160715133437] [PMID: 27426126]
[30]
Azziz, R. PCOS in 2015: New insights into the genetics of polycystic ovary syndrome. Nat. Rev. Endocrinol., 2016, 12(3), 183.
[http://dx.doi.org/10.1038/nrendo.2016.9] [PMID: 26822926]
[31]
Dunaif, A. Perspectives in polycystic ovary syndrome: from hair to eternity. J. Clin. Endocrinol. Metab., 2016, 101(3), 759-768.
[http://dx.doi.org/10.1210/jc.2015-3780] [PMID: 26908109]
[32]
Ambros, V. microRNAs: tiny regulators with great potential. Cell, 2001, 107(7), 823-826.
[http://dx.doi.org/10.1016/S0092-8674(01)00616-X] [PMID: 11779458]
[33]
Gallo, A.; Tandon, M.; Alevizos, I.; Illei, G.G. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One, 2012, 7(3)e30679
[http://dx.doi.org/10.1371/journal.pone.0030679] [PMID: 22427800]
[34]
Lawrie, C.H.; Gal, S.; Dunlop, H.M.; Pushkaran, B.; Liggins, A.P.; Pulford, K.; Banham, A.H.; Pezzella, F.; Boultwood, J.; Wainscoat, J.S.; Hatton, C.S.; Harris, A.L. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br. J. Haematol., 2008, 141(5), 672-675.
[http://dx.doi.org/10.1111/j.1365-2141.2008.07077.x] [PMID: 18318758]
[35]
Hanson, E.K.; Lubenow, H.; Ballantyne, J. Identification of forensically relevant body fluids using a panel of differentially expressed microRNAs. Anal. Biochem., 2009, 387(2), 303-314.
[http://dx.doi.org/10.1016/j.ab.2009.01.037] [PMID: 19454234]
[36]
Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; Lin, D.W.; Urban, N.; Drescher, C.W.; Knudsen, B.S.; Stirewalt, D.L.; Gentleman, R.; Vessella, R.L.; Nelson, P.S.; Martin, D.B.; Tewari, M. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA, 2008, 105(30), 10513-10518.
[http://dx.doi.org/10.1073/pnas.0804549105] [PMID: 18663219]
[37]
Osman, A. MicroRNAs in health and disease--basic science and clinical applications. Clin. Lab., 2012, 58(5-6), 393-402.
[PMID: 22783567]
[38]
Fernandez-Valverde, S.L.; Taft, R.J.; Mattick, J.S. MicroRNAs in β-cell biology, insulin resistance, diabetes and its complications. Diabetes, 2011, 60(7), 1825-1831.
[http://dx.doi.org/10.2337/db11-0171] [PMID: 21709277]
[39]
Hulsmans, M.; De Keyzer, D.; Holvoet, P. MicroRNAs regulating oxidative stress and inflammation in relation to obesity and atherosclerosis. FASEB J., 2011, 25(8), 2515-2527.
[http://dx.doi.org/10.1096/fj.11-181149] [PMID: 21507901]
[40]
Carletti, M.Z.; Fiedler, S.D.; Christenson, L.K. MicroRNA 21 blocks apoptosis in mouse periovulatory granulosa cells. Biol. Reprod., 2010, 83(2), 286-295.
[http://dx.doi.org/10.1095/biolreprod.109.081448] [PMID: 20357270]
[41]
Ding, C-F.; Chen, W-Q.; Zhu, Y-T.; Bo, Y-L.; Hu, H-M.; Zheng, R-H. Circulating microRNAs in patients with polycystic ovary syndrome. Hum. Fertil. (Camb.), 2015, 18(1), 22-29.
[http://dx.doi.org/10.3109/14647273.2014.956811] [PMID: 25268995]
[42]
Long, W.; Zhao, C.; Ji, C.; Ding, H.; Cui, Y.; Guo, X.; Shen, R.; Liu, J. Characterization of serum microRNAs profile of PCOS and identification of novel non-invasive biomarkers. Cell. Physiol. Biochem., 2014, 33(5), 1304-1315.
[http://dx.doi.org/10.1159/000358698] [PMID: 24802714]
[43]
Murri, M.; Insenser, M.; Fernández-Durán, E.; San-Millán, J.L.; Escobar-Morreale, H.F. Effects of polycystic ovary syndrome (PCOS), sex hormones and obesity on circulating miRNA-21, miRNA-27b, miRNA-103, and miRNA-155 expression. J. Clin. Endocrinol. Metab., 2013, 98(11), E1835-E1844.
[http://dx.doi.org/10.1210/jc.2013-2218] [PMID: 24037889]
[44]
Naji, M.; Aleyasin, A.; Nekoonam, S.; Arefian, E.; Mahdian, R.; Amidi, F. Differential expression of miR-93 and miR-21 in granulosa cells and follicular fluid of polycystic ovary syndrome associating with different phenotypes. Sci. Rep., 2017, 7(1), 14671.
[http://dx.doi.org/10.1038/s41598-017-13250-1] [PMID: 29116087]
[45]
Rau, C-S.; Yang, J.C.; Wu, S.C.; Chen, Y.C.; Lu, T.H.; Lin, M.W.; Wu, Y.C.; Tzeng, S.L.; Wu, C.J.; Hsieh, C.H. Profiling circulating microRNA expression in a mouse model of nerve allotransplantation. J. Biomed. Sci., 2013, 20, 64.
[http://dx.doi.org/10.1186/1423-0127-20-64] [PMID: 24011263]
[46]
Roth, L.W.; McCallie, B.; Alvero, R.; Schoolcraft, W.B.; Minjarez, D.; Katz-Jaffe, M.G. Altered microRNA and gene expression in the follicular fluid of women with polycystic ovary syndrome. J. Assist. Reprod. Genet., 2014, 31(3), 355-362.
[http://dx.doi.org/10.1007/s10815-013-0161-4] [PMID: 24390626]
[47]
Salimi-Asl, M.; Mozdarani, H.; Kadivar, M. Up-regulation of miR-21 and 146a expression and increased DNA damage frequency in a mouse model of polycystic ovary syndrome (PCOS). Bioimpacts, 2016, 6(2), 85-91.
[http://dx.doi.org/10.15171/bi.2016.12] [PMID: 27525225]
[48]
Sang, Q.; Yao, Z.; Wang, H.; Feng, R.; Wang, H.; Zhao, X.; Xing, Q.; Jin, L.; He, L.; Wu, L.; Wang, L. Identification of microRNAs in human follicular fluid: characterization of microRNAs that govern steroidogenesis in vitro and are associated with polycystic ovary syndrome in vivo. J. Clin. Endocrinol. Metab., 2013, 98(7), 3068-3079.
[http://dx.doi.org/10.1210/jc.2013-1715] [PMID: 23666971]
[49]
Scalici, E.; Traver, S.; Mullet, T.; Molinari, N.; Ferrières, A.; Brunet, C.; Belloc, S.; Hamamah, S. Circulating microRNAs in follicular fluid, powerful tools to explore in vitro fertilization process. Sci. Rep., 2016, 6, 24976.
[http://dx.doi.org/10.1038/srep24976] [PMID: 27102646]
[50]
Sirotkin, A.V.; Ovcharenko, D.; Grossmann, R.; Lauková, M.; Mlyncek, M. Identification of microRNAs controlling human ovarian cell steroidogenesis via a genome-scale screen. J. Cell. Physiol., 2009, 219(2), 415-420.
[http://dx.doi.org/10.1002/jcp.21689] [PMID: 19194990]
[51]
Sørensen, A.E.; Wissing, M.L.; Salö, S.; Englund, A.L.M.; Dalgaard, L.T. MicroRNAs related to polycystic ovary syndrome (PCOS). Genes (Basel), 2014, 5(3), 684-708.
[http://dx.doi.org/10.3390/genes5030684] [PMID: 25158044]
[52]
Sørensen, A.E.; Udesen, P.B.; Wissing, M.L.; Englund, A.L.M.; Dalgaard, L.T. MicroRNAs related to androgen metabolism and polycystic ovary syndrome. Chem. Biol. Interact., 2016, 259(Pt A), 8-16.
[http://dx.doi.org/10.1016/j.cbi.2016.06.008] [PMID: 27270454]
[53]
Yin, M.; Wang, X.; Yao, G.; Lü, M.; Liang, M.; Sun, Y.; Sun, F. Transactivation of micrornA-320 by microRNA-383 regulates granulosa cell functions by targeting E2F1 and SF-1 proteins. J. Biol. Chem., 2014, 289(26), 18239-18257.
[http://dx.doi.org/10.1074/jbc.M113.546044] [PMID: 24828505]
[54]
Zhong, Z.; Li, F.; Li, Y.; Qin, S.; Wen, C.; Fu, Y.; Xiao, Q. Inhibition of microRNA-19b promotes ovarian granulosa cell proliferation by targeting IGF-1 in polycystic ovary syndrome. Mol. Med. Rep., 2018, 17(4), 4889-4898.
[http://dx.doi.org/10.3892/mmr.2018.8463] [PMID: 29363717]
[55]
Li, C.; Chen, L.; Zhao, Y.; Chen, S.; Fu, L.; Jiang, Y.; Gao, S.; Liu, Z.; Wang, F.; Zhu, X.; Rao, J.; Zhang, J.; Zhou, X. Altered expression of miRNAs in the uterus from a letrozole-induced rat PCOS model. Gene, 2017, 598, 20-26.
[http://dx.doi.org/10.1016/j.gene.2016.10.033] [PMID: 27777110]
[56]
Hossain, M.M.; Cao, M.; Wang, Q.; Kim, J.Y.; Schellander, K.; Tesfaye, D.; Tsang, B.K. Altered expression of miRNAs in a dihydrotestosterone-induced rat PCOS model. J. Ovarian Res., 2013, 6(1), 36.
[http://dx.doi.org/10.1186/1757-2215-6-36] [PMID: 23675970]
[57]
Chang, R.J.; Cook-Andersen, H. Disordered follicle development. Mol. Cell. Endocrinol., 2013, 373(1-2), 51-60.
[http://dx.doi.org/10.1016/j.mce.2012.07.011] [PMID: 22874072]
[58]
Sirotkin, A.V.; Lauková, M.; Ovcharenko, D.; Brenaut, P.; Mlyncek, M. Identification of microRNAs controlling human ovarian cell proliferation and apoptosis. J. Cell. Physiol., 2010, 223(1), 49-56.
[http://dx.doi.org/10.1002/jcp.21999 ] [PMID: 20039279]
[59]
Lu, J.; Zhang, C.; Gu, B.; Zhang, S.; Geng, J.; Chen, Y.; Xie, J. 2017.
[60]
Geng, Y.; Sui, C.; Xun, Y.; Lai, Q.; Jin, L. MiRNA-99a can regulate proliferation and apoptosis of human granulosa cells via targeting IGF-1R in polycystic ovary syndrome. J. Assist. Reprod. Genet., 2019, 36(2), 211-221.
[http://dx.doi.org/10.1007/s10815-018-1335-x ] [PMID: 30374732]
[61]
Wang, T.; Liu, Y.; Lv, M.; Xing, Q.; Zhang, Z.; He, X.; Xu, Y.; Wei, Z.; Cao, Y. miR-323-3p regulates the steroidogenesis and cell apoptosis in polycystic ovary syndrome (PCOS) by targeting IGF-1. Gene, 2019, 683, 87-100.
[http://dx.doi.org/10.1016/j.gene.2018.10.006] [PMID: 30300681]
[62]
Zhang, Z.; Chen, C.Z.; Xu, M.Q.; Zhang, L.Q.; Liu, J.B.; Gao, Y.; Jiang, H.; Yuan, B.; Zhang, J.B. MiR-31 and miR-143 affect steroid hormone synthesis and inhibit cell apoptosis in bovine granulosa cells through FSHR. Theriogenology, 2019, 123, 45-53.
[http://dx.doi.org/10.1016/j.theriogenology.2018.09.020] [PMID: 30278258]
[63]
Sun, X-F.; Li, Y-P.; Pan, B.; Wang, Y-F.; Li, J.; Shen, W. Molecular regulation of miR-378 on the development of mouse follicle and the maturation of oocyte in vivo. Cell Cycle, 2018, 17(18), 2230-2242.
[http://dx.doi.org/10.1080/15384101.2018.1520557] [PMID: 30244637]
[64]
Fu, X.; He, Y.; Wang, X.; Peng, D.; Chen, X.; Li, X.; Wan, Q. MicroRNA-16 promotes ovarian granulosa cell proliferation and suppresses apoptosis through targeting PDCD4 in polycystic ovarian syndrome. Cell. Physiol. Biochem., 2018, 48(2), 670-682.
[http://dx.doi.org/10.1159/000491894] [PMID: 30025387]
[65]
Jiang, Y-C.; Ma, J-X. The role of MiR-324-3p in polycystic ovary syndrome (PCOS) via targeting WNT2B. Eur. Rev. Med. Pharmacol. Sci., 2018, 22(11), 3286-3293.
[http://dx.doi.org/10.26355/eurrev_201806_15147 ] [PMID: 29917177]
[66]
Liu, J.; Li, X.; Yao, Y.; Li, Q.; Pan, Z.; Li, Q. miR-1275 controls granulosa cell apoptosis and estradiol synthesis by impairing LRH-1/CYP19A1 axis. Biochim. Biophys. Acta. Gene Regul. Mech., 2018, 1861(3), 246-257.
[http://dx.doi.org/10.1016/j.bbagrm.2018.01.009] [PMID: 29378329]
[67]
Wang, M.; Liu, M.; Sun, J.; Jia, L.; Ma, S.; Gao, J.; Xu, Y.; Zhang, H.; Tsang, S.Y.; Li, X. MicroRNA-27a-3p affects estradiol and androgen imbalance by targeting Creb1 in the granulosa cells in mouse polycytic ovary syndrome model. Reprod. Biol., 2017, 17(4), 295-304.
[http://dx.doi.org/10.1016/j.repbio.2017.09.005] [PMID: 29089199]
[68]
Wang, M.; Sun, J.; Xu, B.; Chrusciel, M.; Gao, J.; Bazert, M.; Stelmaszewska, J.; Xu, Y.; Zhang, H.; Pawelczyk, L.; Sun, F.; Ying, S.; Rahman, N.; Wołczyński, S.; Li, X. Functional characterization of MicroRNA-27a-3p expression in human polycystic ovary syndrome. Endocrinology, 2018, 159(1), 297-309.
[http://dx.doi.org/10.1210/en.2017-00219]]
[69]
Yang, L.; Li, Y.; Wang, X.; Liu, Y.; Yang, L. MicroRNA320a inhibition decreases insulininduced KGN cell proliferation and apoptosis by targeting PCGF1. Mol. Med. Rep., 2017, 16(4), 5706-5712.
[http://dx.doi.org/10.3892/mmr.2017.7270] [PMID: 28849208]
[70]
Mao, Z.; Fan, L.; Yu, Q.; Luo, S.; Wu, X.; Tang, J.; Kang, G.; Tang, L. Abnormality of klotho signaling is involved in polycystic ovary syndrome. Reprod. Sci., 2018, 25(3), 372-383.
[http://dx.doi.org/10.1177/1933719117715129] [PMID: 28673204]
[71]
Li, D.; Xu, D.; Xu, Y.; Chen, L.; Li, C.; Dai, X.; Zhang, L.; Zheng, L. MicroRNA-141-3p targets DAPK1 and inhibits apoptosis in rat ovarian granulosa cells. Cell Biochem. Funct., 2017, 35(4), 197-201.
[http://dx.doi.org/10.1002/cbf.3248] [PMID: 28543175]
[72]
Cai, G.; Ma, X.; Chen, B.; Huang, Y.; Liu, S.; Yang, H.; Zou, W. MicroRNA-145 negatively regulates cell proliferation through targeting IRS1 in isolated ovarian granulosa cells from patients with polycystic ovary syndrome. Reprod. Sci., 2017, 24(6), 902-910.
[http://dx.doi.org/10.1177/1933719116673197] [PMID: 27799458]
[73]
Tiwari, M.; Prasad, S.; Tripathi, A.; Pandey, A.N.; Ali, I.; Singh, A.K.; Shrivastav, T.G.; Chaube, S.K. Apoptosis in mammalian oocytes: a review. Apoptosis, 2015, 20(8), 1019-1025.
[http://dx.doi.org/10.1007/s10495-015-1136-y] [PMID: 25958165]
[74]
Luo, H.; Han, Y.; Liu, J.; Zhang, Y. Identification of microRNAs in granulosa cells from patients with different levels of ovarian reserve function and the potential regulatory function of miR-23a in granulosa cell apoptosis. Gene, 2019, 686, 250-260.
[http://dx.doi.org/10.1016/j.gene.2018.11.025] [PMID: 30453069]
[75]
Yao, Y.; Niu, J.; Sizhu, S.; Li, B.; Chen, Y.; Li, R.; Yangzong, Q.; Li, Q.; Xu, Y. microRNA-125b regulates apoptosis by targeting bone morphogenetic protein receptor 1B in yak granulosa cells. DNA Cell Biol., 2018, 37(11), 878-887.
[http://dx.doi.org/10.1089/dna.2018.4354] [PMID: 30260685]
[76]
Zhang, H.; Jiang, X.; Zhang, Y.; Xu, B.; Hua, J.; Ma, T.; Zheng, W.; Sun, R.; Shen, W.; Cooke, H.J.; Hao, Q.; Qiao, J.; Shi, Q. microRNA 376a regulates follicle assembly by targeting Pcna in fetal and neonatal mouse ovaries. Reproduction, 2014, 148(1), 43-54.
[http://dx.doi.org/10.1530/REP-13-0508] [PMID: 24686458]
[77]
Zhang, J.; Ji, X.; Zhou, D.; Li, Y.; Lin, J.; Liu, J.; Luo, H.; Cui, S. miR-143 is critical for the formation of primordial follicles in mice. Front. Biosci., 2013, 18, 588-597.
[http://dx.doi.org/10.2741/4122] [PMID: 23276944]
[78]
Bird, A. Perceptions of epigenetics. Nature, 2007, 447(7143), 396-398.
[http://dx.doi.org/10.1038/nature05913] [PMID: 17522671]
[79]
Guo, F.; Li, X.; Liang, D.; Li, T.; Zhu, P.; Guo, H.; Wu, X.; Wen, L.; Gu, T.P.; Hu, B.; Walsh, C.P.; Li, J.; Tang, F.; Xu, G.L. Active and passive demethylation of male and female pronuclear DNA in the mammalian zygote. Cell Stem Cell, 2014, 15(4), 447-459.
[http://dx.doi.org/10.1016/j.stem.2014.08.003] [PMID: 25220291]
[80]
Qu, F.; Wang, F.F.; Yin, R.; Ding, G.L.; El-Prince, M.; Gao, Q.; Shi, B.W.; Pan, H.H.; Huang, Y.T.; Jin, M.; Leung, P.C.; Sheng, J.Z.; Huang, H.F. A molecular mechanism underlying ovarian dysfunction of polycystic ovary syndrome: hyperandrogenism induces epigenetic alterations in the granulosa cells. J. Mol. Med. (Berl.), 2012, 90(8), 911-923.
[http://dx.doi.org/10.1007/s00109-012-0881-4] [PMID: 22349439]
[81]
Yu, Y-Y.; Sun, C-X.; Liu, Y-K.; Li, Y.; Wang, L.; Zhang, W. Promoter methylation of CYP19A1 gene in Chinese polycystic ovary syndrome patients. Gynecol. Obstet. Invest., 2013, 76(4), 209-213.
[http://dx.doi.org/10.1159/000355314] [PMID: 24157654]
[82]
Wang, P.; Zhao, H.; Li, T.; Zhang, W.; Wu, K.; Li, M.; Bian, Y.; Liu, H.; Ning, Y.; Li, G.; Chen, Z.J. Hypomethylation of the LH/choriogonadotropin receptor promoter region is a potential mechanism underlying susceptibility to polycystic ovary syndrome. Endocrinology, 2014, 155(4), 1445-1452.
[http://dx.doi.org/10.1210/en.2013-1764] [PMID: 24527662]
[83]
Sang, Q.; Li, X.; Wang, H.; Wang, H.; Zhang, S.; Feng, R.; Xu, Y.; Li, Q.; Zhao, X.; Xing, Q.; Jin, L.; He, L.; Wang, L. Quantitative methylation level of the EPHX1 promoter in peripheral blood DNA is associated with polycystic ovary syndrome. PLoS One, 2014, 9(2)e88013
[http://dx.doi.org/10.1371/journal.pone.0088013] [PMID: 24505354]
[84]
Wang, X-X.; Wei, J-Z.; Jiao, J.; Jiang, S-Y.; Yu, D-H.; Li, D. Genome-wide DNA methylation and gene expression patterns provide insight into polycystic ovary syndrome development. Oncotarget, 2014, 5(16), 6603-6610.
[http://dx.doi.org/10.18632/oncotarget.2224] [PMID: 25051372]
[85]
Pan, J-X.; Tan, Y.J.; Wang, F.F.; Hou, N.N.; Xiang, Y.Q.; Zhang, J.Y.; Liu, Y.; Qu, F.; Meng, Q.; Xu, J.; Sheng, J.Z.; Huang, H.F. Aberrant expression and DNA methylation of lipid metabolism genes in PCOS: a new insight into its pathogenesis. Clin. Epigenetics, 2018, 10, 6.
[http://dx.doi.org/10.1186/s13148-018-0442-y] [PMID: 29344314]
[86]
Zhao, H.; Zhao, Y.; Ren, Y.; Li, M.; Li, T.; Li, R.; Yu, Y.; Qiao, J. Epigenetic regulation of an adverse metabolic phenotype in polycystic ovary syndrome: the impact of the leukocyte methylation of PPARGC1A promoter. Fertil. Steril., 2017, 107(2), 467-474.e5.
[http://dx.doi.org/10.1016/j.fertnstert.2016.10.039] [PMID: 27889100]
[87]
Li, Q-N.; Guo, L.; Hou, Y.; Ou, X-H.; Liu, Z.; Sun, Q-Y. The DNA methylation profile of oocytes in mice with hyperinsulinaemia and hyperandrogenism as detected by single-cell level whole genome bisulphite sequencing (SC-WGBS) technology. Reprod. Fertil. Dev., 2018, 30(12), 1713-1719.
[http://dx.doi.org/10.1071/RD18002] [PMID: 29929576]
[88]
Pruksananonda, K.; Wasinarom, A.; Sereepapong, W.; Sirayapiwat, P.; Rattanatanyong, P.; Mutirangura, A. Epigenetic modification of long interspersed elements-1 in cumulus cells of mature and immature oocytes from patients with polycystic ovary syndrome. Clin. Exp. Reprod. Med., 2016, 43(2), 82-89.
[http://dx.doi.org/10.5653/cerm.2016.43.2.82] [PMID: 27358825]
[89]
Salehi Jahromi, M.; Hill, J.W.; Ramezani Tehrani, F.; Zadeh-Vakili, A. Hypomethylation of specific CpG sites in the promoter region of steroidogeneic genes (GATA6 and StAR) in prenatally androgenized rats. Life Sci., 2018, 207, 105-109.
[http://dx.doi.org/10.1016/j.lfs.2018.05.052] [PMID: 29859221]
[90]
Desai, V.; Prasad, N.R.; Manohar, S.M.; Sachan, A.; Narasimha, S.R.P.V.L.; Bitla, A.R.R. Oxidative stress in non-obese women with polycystic ovarian syndrome. J. Clin. Diagn. Res., 2014, 8(7), CC01-CC03.
[http://dx.doi.org/10.7860/JCDR/2014/8125.4530 ] [PMID: 25177558]
[91]
Elia, E.M.; Belgorosky, D.; Faut, M.; Vighi, S.; Pustovrh, C.; Luigi, D.; Motta, A.B. The effects of metformin on uterine tissue of hyperandrogenized BALB/c mice. Mol. Hum. Reprod., 2009, 15(7), 421-432.
[http://dx.doi.org/10.1093/molehr/gap033] [PMID: 19482906]
[92]
Luchetti, C.G.; Mikó, E.; Szekeres-Bartho, J.; Paz, D.A.; Motta, A.B. Dehydroepiandrosterone and metformin modulate progesterone-induced blocking factor (PIBF), cyclooxygenase 2 (COX2) and cytokines in early pregnant mice. J. Steroid Biochem. Mol. Biol., 2008, 111(3-5), 200-207.
[http://dx.doi.org/10.1016/j.jsbmb.2008.06.007] [PMID: 18606228]
[93]
Rocha, M.; Diaz-Morales, N.; Rovira-Llopis, S.; Escribano-Lopez, I.; Bañuls, C.; Hernandez-Mijares, A.; Diamanti-Kandarakis, E.; Victor, V.M. Mitochondrial dysfunction and endoplasmic reticulum stress in diabetes. Curr. Pharm. Des., 2016, 22(18), 2640-2649.
[http://dx.doi.org/10.2174/1381612822666160209152033] [PMID: 26861650]
[94]
Zuo, T.; Zhu, M.; Xu, W. Roles of oxidative stress in polycystic ovary syndrome and cancers. Oxid. Med. Cell. Longev., 2016, 20168589318
[http://dx.doi.org/10.1155/2016/8589318] [PMID: 26770659]
[95]
Amalfi, S.; Velez, L.M.; Heber, M.F.; Vighi, S.; Ferreira, S.R.; Orozco, A.V.; Pignataro, O.; Motta, A.B. Prenatal hyperandrogenization induces metabolic and endocrine alterations which depend on the levels of testosterone exposure. PLoS One, 2012, 7(5)e37658
[http://dx.doi.org/10.1371/journal.pone.0037658] [PMID: 22655062]
[96]
Motta, A.B. Editorial (thematic issue: advances in the diagnosis and treatment of polycystic ovarian syndrome). Curr. Pharm. Des., 2016, 22(36), 5505-5507.
[http://dx.doi.org/10.2174/1381612822666160826121954] [PMID: 27568785]
[97]
Eini, F.; Novin, M.G.; Joharchi, K.; Hosseini, A.; Nazarian, H.; Piryaei, A.; Bidadkosh, A. Intracytoplasmic oxidative stress reverses epigenetic modifications in polycystic ovary syndrome. Reprod. Fertil. Dev., 2017, 29(12), 2313-2323.
[http://dx.doi.org/10.1071/RD16428] [PMID: 28442024]
[98]
Salehi Jahromi, M.; Ramezani Tehrani, F.; Hill, J.W.; Noroozzadeh, M.; Zarkesh, M.; Ghasemi, A.; Zadeh-Vakili, A. Alteration in follistatin gene expression detected in prenatally androgenized rats. Gynecol. Endocrinol., 2017, 33(6), 433-437.
[http://dx.doi.org/10.1080/09513590.2017.1290067] [PMID: 28277126]
[99]
Yi, L.; Huang, X.; Guo, F.; Zhou, Z.; Dou, Y.; Huan, J. Yes-associated protein (YAP) signaling regulates lipopolysaccharide-induced tissue factor expression in human endothelial cells. Surgery, 2016, 159(5), 1436-1448.
[http://dx.doi.org/10.1016/j.surg.2015.12.008] [PMID: 26791271]
[100]
Jiang, L-L.; Xie, J.K.; Cui, J.Q.; Wei, D.; Yin, B.L.; Zhang, Y.N.; Chen, Y.H.; Han, X.; Wang, Q.; Zhang, C.L. Promoter methylation of yes-associated protein (YAP1) gene in polycystic ovary syndrome. Medicine (Baltimore), 2017, 96(2)e5768
[http://dx.doi.org/10.1097/MD.0000000000005768] [PMID: 28079802]
[101]
Li, S.; Zhu, D.; Duan, H.; Ren, A.; Glintborg, D.; Andersen, M.; Skov, V.; Thomassen, M.; Kruse, T.; Tan, Q. Differential DNA methylation patterns of polycystic ovarian syndrome in whole blood of Chinese women. Oncotarget, 2017, 8(13), 20656-20666.
[http://dx.doi.org/10.18632/oncotarget.9327] [PMID: 27192117]
[102]
Mannerås-Holm, L.; Benrick, A.; Stener-Victorin, E. Gene expression in subcutaneous adipose tissue differs in women with polycystic ovary syndrome and controls matched pair-wise for age, body weight, and body mass index. Adipocyte, 2014, 3(3), 190-196.
[http://dx.doi.org/10.4161/adip.28731] [PMID: 25068085]
[103]
Kokosar, M.; Benrick, A.; Perfilyev, A.; Fornes, R.; Nilsson, E.; Maliqueo, M.; Behre, C.J.; Sazonova, A.; Ohlsson, C.; Ling, C.; Stener-Victorin, E. Epigenetic and transcriptional alterations in human adipose tissue of polycystic ovary syndrome. Sci. Rep., 2016, 6, 22883.
[http://dx.doi.org/10.1038/srep22883] [PMID: 26975253]
[104]
Saenz-de-Juano, M.D.; Billooye, K.; Smitz, J.; Anckaert, E. The loss of imprinted DNA methylation in mouse blastocysts is inflicted to a similar extent by in vitro follicle culture and ovulation induction. Mol. Hum. Reprod., 2016, 22(6), 427-441.
[http://dx.doi.org/10.1093/molehr/gaw013] [PMID: 26908643]
[105]
Zhu, J-Q.; Zhu, L.; Liang, X-W.; Xing, F-Q.; Schatten, H.; Sun, Q-Y. Demethylation of LHR in dehydroepiandrosterone-induced mouse model of polycystic ovary syndrome. Mol. Hum. Reprod., 2010, 16(4), 260-266.
[http://dx.doi.org/10.1093/molehr/gap089] [PMID: 19828691]
[106]
Arancio, W.; Calogero Amato, M.; Magliozzo, M.; Pizzolanti, G.; Vesco, R.; Giordano, C. Serum miRNAs in women affected by hyperandrogenic polycystic ovary syndrome: the potential role of miR-155 as a biomarker for monitoring the estroprogestinic treatment. Gynecol. Endocrinol., 2018, 34(8), 704-708.
[http://dx.doi.org/10.1080/09513590.2018.1428299] [PMID: 29385860]
[107]
Zhai, J. Metformin regulates key MicroRNAs to improve endome-trial receptivity through increasing implantation marker gene expression in patients with PCOS undergoing IVF/ICSI. Reprod. Sci. Thousand Oaks Calif, 2019, 26(1)193371911882046
[http://dx.doi.org/10.1177/1933719118820466 ]
[108]
Zhao, H.; Zhou, D.; Chen, Y.; Liu, D.; Chu, S.; Zhang, S. Beneficial effects of Heqi san on rat model of polycystic ovary syndrome through the PI3K/AKT pathway. Daru, 2017, 25(1), 21.
[http://dx.doi.org/10.1186/s40199-017-0188-7] [PMID: 29020999]
[109]
Christopher, A.F.; Kaur, R.P.; Kaur, G.; Kaur, A.; Gupta, V.; Bansal, P. MicroRNA therapeutics: discovering novel targets and developing specific therapy. Perspect. Clin. Res., 2016, 7(2), 68-74.
[http://dx.doi.org/10.4103/2229-3485.179431] [PMID: 27141472]
[110]
Chen, B.; Ye, F.; Yu, L.; Jia, G.; Huang, X.; Zhang, X.; Peng, S.; Chen, K.; Wang, M.; Gong, S.; Zhang, R.; Yin, J.; Li, H.; Yang, Y.; Liu, H.; Zhang, J.; Zhang, H.; Zhang, A.; Jiang, H.; Luo, C.; Yang, C.G. Development of cell-active N6-methyladenosine RNA demethylase FTO inhibitor. J. Am. Chem. Soc., 2012, 134(43), 17963-17971.
[http://dx.doi.org/10.1021/ja3064149] [PMID: 23045983]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy