DOI QR코드

DOI QR Code

Lifetime changes of the oocyte pool: Contributing factors with a focus on ovulatory inflammation

  • Park, Chan Jin (Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign) ;
  • Oh, Ji-Eun (Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign) ;
  • Feng, Jianan (Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign) ;
  • Cho, Yoon Min (Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign) ;
  • Qiao, Huanyu (Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign) ;
  • Ko, CheMyong (Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign)
  • Received : 2021.09.03
  • Accepted : 2022.02.11
  • Published : 2022.03.31

Abstract

In mammalian species, females are born with a number of oocytes exceeding what they release via ovulation. In humans, an average girl is born with over a thousand times more oocytes than she will ovulate in her lifetime. The reason for having such an excessive number of oocytes in a neonatal female ovary is currently unknown. However, it is well established that the oocyte number decreases throughout the entire lifetime until the ovary loses them all. In this review, data published in the past 80 years were used to assess the current knowledge regarding the changing number of oocytes in humans and mice, as well as the reported factors that contribute to the decline of oocyte numbers. Briefly, a collective estimation indicates that an average girl is born with approximately 600,000 oocytes, which is 2,000 times more than the number of oocytes that she will ovulate in her lifetime. The oocyte number begins to decrease immediately after birth and is reduced to half of the initial number by puberty and almost zero by age 50 years. Multiple factors that are either intrinsic or extrinsic to the ovary contribute to the decline of the oocyte number. The inflammation caused by the ovulatory luteinizing hormone surge is discussed as a potential contributing factor to the decline of the oocyte pool during the reproductive lifespan.

Keywords

Acknowledgement

This study was supported by NIH HD094296 (CK), R00 HD082375 (HQ), and NSF SBIR 2052603 (CP).

References

  1. Pastore LM, Christianson MS, Stelling J, Kearns WG, Segars JH. Reproductive ovarian testing and the alphabet soup of diagnoses: DOR, POI, POF, POR, and FOR. J Assist Reprod Genet 2018;35:17-23. https://doi.org/10.1007/s10815-017-1058-4
  2. Block E. Quantitative morphological investigations of the follicular system in women; variations at different ages. Acta Anat (Basel) 1952;14:108-23. https://doi.org/10.1159/000140595
  3. Block E. A quantitative morphological investigation of the follicular system in newborn female infants. Acta Anat (Basel) 1953;17:201-6. https://doi.org/10.1159/000140805
  4. Faddy MJ, Gosden RG. A mathematical model of follicle dynamics in the human ovary. Hum Reprod 1995;10:770-5. https://doi.org/10.1093/oxfordjournals.humrep.a136036
  5. Forabosco A, Sforza C. Establishment of ovarian reserve: a quantitative morphometric study of the developing human ovary. Fertil Steril 2007;88:675-83. https://doi.org/10.1016/j.fertnstert.2006.11.191
  6. Gougeon A, Ecochard R, Thalabard JC. Age-related changes of the population of human ovarian follicles: increase in the disappearance rate of non-growing and early-growing follicles in aging women. Biol Reprod 1994;50:653-63. https://doi.org/10.1095/biolreprod50.3.653
  7. Hansen KR, Knowlton NS, Thyer AC, Charleston JS, Soules MR, Klein NA. A new model of reproductive aging: the decline in ovarian non-growing follicle number from birth to menopause. Hum Reprod 2008;23:699-708. https://doi.org/10.1093/humrep/dem408
  8. Richardson SJ, Senikas V, Nelson JF. Follicular depletion during the menopausal transition: evidence for accelerated loss and ultimate exhaustion. J Clin Endocrinol Metab 1987;65:1231-7. https://doi.org/10.1210/jcem-65-6-1231
  9. Sonne-Hansen K, Nielsen M, Byskov AG. Oocyte number in newborn mice after prenatal octylphenol exposure. Reprod Toxicol 2003;17:59-66. https://doi.org/10.1016/S0890-6238(02)00079-5
  10. Pepling ME, Spradling AC. Mouse ovarian germ cell cysts undergo programmed breakdown to form primordial follicles. Dev Biol 2001;234:339-51. https://doi.org/10.1006/dbio.2001.0269
  11. Danilovich N, Sairam MR. Haploinsufficiency of the follicle-stimulating hormone receptor accelerates oocyte loss inducing early reproductive senescence and biological aging in mice. Biol Reprod 2002;67:361-9. https://doi.org/10.1095/biolreprod67.2.361
  12. Fu X, Cheng J, Hou Y, Zhu S. The association between the oocyte pool and aneuploidy: a comparative study of the reproductive potential of young and aged mice. J Assist Reprod Genet 2014;31:323-31. https://doi.org/10.1007/s10815-013-0160-5
  13. Kerr JB, Duckett R, Myers M, Britt KL, Mladenovska T, Findlay JK. Quantification of healthy follicles in the neonatal and adult mouse ovary: evidence for maintenance of primordial follicle supply. Reproduction 2006;132:95-109. https://doi.org/10.1530/rep.1.01128
  14. Uri-Belapolsky S, Shaish A, Eliyahu E, Grossman H, Levi M, Chuderland D, et al. Interleukin-1 deficiency prolongs ovarian lifespan in mice. Proc Natl Acad Sci U S A 2014;111:12492-7. https://doi.org/10.1073/pnas.1323955111
  15. Skaznik-Wikiel M, Tilly JC, Lee HJ, Niikura Y, Kaneko-Tarui T, Johnson J, et al. Serious doubts over "Eggs forever?". Differentiation 2007;75:93-9. https://doi.org/10.1111/j.1432-0436.2006.00117.x
  16. Johnson J, Canning J, Kaneko T, Pru JK, Tilly JL. Germline stem cells and follicular renewal in the postnatal mammalian ovary. Nature 2004;428:145-50. https://doi.org/10.1038/nature02316
  17. Tamura H, Kawamoto M, Sato S, Tamura I, Maekawa R, Taketani T, et al. Long-term melatonin treatment delays ovarian aging. J Pineal Res 2017;62:e12381. https://doi.org/10.1111/jpi.12381
  18. Miao Y, Cui Z, Gao Q, Rui R, Xiong B. Nicotinamide mononucleotide supplementation reverses the declining quality of maternally aged oocytes. Cell Rep 2020;32:107987. https://doi.org/10.1016/j.celrep.2020.107987
  19. Buyuk E, Seifer DB, Younger J, Grazi RV, Lieman H. Random anti-Mullerian hormone (AMH) is a predictor of ovarian response in women with elevated baseline early follicular follicle-stimulating hormone levels. Fertil Steril 2011;95:2369-72. https://doi.org/10.1016/j.fertnstert.2011.03.071
  20. Weghofer A, Barad DH, Darmon SK, Kushnir VA, Albertini DF, Gleicher N. The ovarian sensitivity index is predictive of live birth chances after IVF in infertile patients. Hum Reprod Open 2020;2020:hoaa049. https://doi.org/10.1093/hropen/hoaa049
  21. Tobler KJ, Shoham G, Christianson MS, Zhao Y, Leong M, Shoham Z. Use of anti-mullerian hormone for testing ovarian reserve: a survey of 796 infertility clinics worldwide. J Assist Reprod Genet 2015;32:1441-8. https://doi.org/10.1007/s10815-015-0562-7
  22. Grive KJ. Pathways coordinating oocyte attrition and abundance during mammalian ovarian reserve establishment. Mol Reprod Dev 2020;87:843-56. https://doi.org/10.1002/mrd.23401
  23. Malki S, van der Heijden GW, O'Donnell KA, Martin SL, Bortvin A. A role for retrotransposon LINE-1 in fetal oocyte attrition in mice. Dev Cell 2014;29:521-33. https://doi.org/10.1016/j.devcel.2014.04.027
  24. Lei L, Spradling AC. Mouse oocytes differentiate through organelle enrichment from sister cyst germ cells. Science 2016;352:95-9. https://doi.org/10.1126/science.aad2156
  25. Kaipia A, Hsueh AJ. Regulation of ovarian follicle atresia. Annu Rev Physiol 1997;59:349-63. https://doi.org/10.1146/annurev.physiol.59.1.349
  26. Hunter N. Oocyte quality control: causes, mechanisms, and consequences. Cold Spring Harb Symp Quant Biol 2017;82:235-47. https://doi.org/10.1101/sqb.2017.82.035394
  27. Qiao H, Rao HB, Yun Y, Sandhu S, Fong JH, Sapre M, et al. Impeding DNA break repair enables oocyte quality control. Mol Cell 2018;72:211-21. https://doi.org/10.1016/j.molcel.2018.08.031
  28. Gawriluk TR, Hale AN, Flaws JA, Dillon CP, Green DR, Rucker EB 3rd. Autophagy is a cell survival program for female germ cells in the murine ovary. Reproduction 2011;141:759-65. https://doi.org/10.1530/REP-10-0489
  29. Song ZH, Yu HY, Wang P, Mao GK, Liu WX, Li MN, et al. Germ cell-specific Atg7 knockout results in primary ovarian insufficiency in female mice. Cell Death Dis 2015;6:e1589. https://doi.org/10.1038/cddis.2014.559
  30. Zhihan T, Xinyi M, Qingying L, Rufei G, Yan Z, Xuemei C, et al. Autophagy participates in cyst breakdown and primordial folliculogenesis by reducing reactive oxygen species levels in perinatal mouse ovaries. J Cell Physiol 2019;234:6125-35. https://doi.org/10.1002/jcp.27367
  31. Wang JJ, Ge W, Zhai QY, Liu JC, Sun XW, Liu WX, et al. Single-cell transcriptome landscape of ovarian cells during primordial follicle assembly in mice. PLoS Biol 2020;18:e3001025. https://doi.org/10.1371/journal.pbio.3001025
  32. Marcinkiewicz JL, Krishna A, Cheung CM, Terranova PF. Oocytic tumor necrosis factor alpha: localization in the neonatal ovary and throughout follicular development in the adult rat. Biol Reprod 1994;50:1251-60. https://doi.org/10.1095/biolreprod50.6.1251
  33. Hatzirodos N, Irving-Rodgers HF, Hummitzsch K, Harland ML, Morris SE, Rodgers RJ. Transcriptome profiling of granulosa cells of bovine ovarian follicles during growth from small to large antral sizes. BMC Genomics 2014;15:24. https://doi.org/10.1186/1471-2164-15-24
  34. Hsueh AJ, Billig H, Tsafriri A. Ovarian follicle atresia: a hormonally controlled apoptotic process. Endocr Rev 1994;15:707-24. https://doi.org/10.1210/edrv-15-6-707
  35. Liu J, Du X, Zhou J, Pan Z, Liu H, Li Q. MicroRNA-26b functions as a proapoptotic factor in porcine follicular Granulosa cells by targeting Sma- and Mad-related protein 4. Biol Reprod 2014;91:146.
  36. Manabe N, Goto Y, Matsuda-Minehata F, Inoue N, Maeda A, Sakamaki K, et al. Regulation mechanism of selective atresia in porcine follicles: regulation of granulosa cell apoptosis during atresia. J Reprod Dev 2004;50:493-514. https://doi.org/10.1262/jrd.50.493
  37. Wigglesworth K, Lee KB, O'Brien MJ, Peng J, Matzuk MM, Eppig JJ. Bidirectional communication between oocytes and ovarian follicular somatic cells is required for meiotic arrest of mammalian oocytes. Proc Natl Acad Sci U S A 2013;110:E3723-9.
  38. Alam MH, Miyano T. Interaction between growing oocytes and granulosa cells in vitro. Reprod Med Biol 2019;19:13-23. https://doi.org/10.1002/rmb2.12292
  39. Alam MH, Lee J, Miyano T. GDF9 and BMP15 induce development of antrum-like structures by bovine granulosa cells without oocytes. J Reprod Dev 2018;64:423-31. https://doi.org/10.1262/jrd.2018-078
  40. Matsuda F, Inoue N, Manabe N, Ohkura S. Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells. J Reprod Dev 2012;58:44-50. https://doi.org/10.1262/jrd.2011-012
  41. Cheng Y, Zhang J, Wu T, Jiang X, Jia H, Qing S, et al. Reproductive toxicity of acute Cd exposure in mouse: resulting in oocyte defects and decreased female fertility. Toxicol Appl Pharmacol 2019;379:114684. https://doi.org/10.1016/j.taap.2019.114684
  42. Li FP, Zhou JL, Guo AW, Liu Y, Zhang F, Xu BH, et al. Di(n-butyl) phthalate exposure impairs meiotic competence and development of mouse oocyte. Environ Pollut 2019;246:597-607. https://doi.org/10.1016/j.envpol.2018.12.077
  43. Xu Y, Sun MH, Xu Y, Ju JQ, Pan MH, Pan ZN, et al. Nonylphenol exposure affects mouse oocyte quality by inducing spindle defects and mitochondria dysfunction. Environ Pollut 2020;266(Pt 1):114967. https://doi.org/10.1016/j.envpol.2020.114967
  44. Goodman RL, Bittman EL, Foster DL, Karsch FJ. The endocrine basis of the synergistic suppression of luteinizing hormone by estradiol and progesterone. Endocrinology 1981;109:1414-7. https://doi.org/10.1210/endo-109-5-1414
  45. Bretveld RW, Thomas CM, Scheepers PT, Zielhuis GA, Roeleveld N. Pesticide exposure: the hormonal function of the female reproductive system disrupted? Reprod Biol Endocrinol 2006;4:30. https://doi.org/10.1186/1477-7827-4-30
  46. Farr SL, Cooper GS, Cai J, Savitz DA, Sandler DP. Pesticide use and menstrual cycle characteristics among premenopausal women in the Agricultural Health Study. Am J Epidemiol 2004;160:1194-204. https://doi.org/10.1093/aje/kwi006
  47. Kalich-Philosoph L, Roness H, Carmely A, Fishel-Bartal M, Ligumsky H, Paglin S, et al. Cyclophosphamide triggers follicle activation and "burnout"; AS101 prevents follicle loss and preserves fertility. Sci Transl Med 2013;5:185ra62. https://doi.org/10.1126/scitranslmed.3005402
  48. Wang Y, Liu M, Johnson SB, Yuan G, Arriba AK, Zubizarreta ME, et al. Doxorubicin obliterates mouse ovarian reserve through both primordial follicle atresia and overactivation. Toxicol Appl Pharmacol 2019;381:114714. https://doi.org/10.1016/j.taap.2019.114714
  49. Morgan S, Anderson RA, Gourley C, Wallace WH, Spears N. How do chemotherapeutic agents damage the ovary? Hum Reprod Update 2012;18:525-35. https://doi.org/10.1093/humupd/dms022
  50. Spears N, Lopes F, Stefansdottir A, Rossi V, De Felici M, Anderson RA, et al. Ovarian damage from chemotherapy and current approaches to its protection. Hum Reprod Update 2019;25:673-93. https://doi.org/10.1093/humupd/dmz027
  51. Winship AL, Carpenter M, Griffiths M, Hutt KJ. Vincristine chemotherapy induces atresia of growing ovarian follicles in mice. Toxicol Sci 2019;169:43-53. https://doi.org/10.1093/toxsci/kfz022
  52. Mishra B, Ripperdan R, Ortiz L, Luderer U. Very low doses of heavy oxygen ion radiation induce premature ovarian failure. Reproduction 2017;154:123-33. https://doi.org/10.1530/REP-17-0101
  53. Malott KF, Luderer U. Toxicant effects on mammalian oocyte mitochondria. Biol Reprod 2021;104:784-93. https://doi.org/10.1093/biolre/ioab002
  54. Lane S, Kauppi L. Meiotic spindle assembly checkpoint and aneuploidy in males versus females. Cell Mol Life Sci 2019;76:1135-50. https://doi.org/10.1007/s00018-018-2986-6
  55. Fragouli E, Wells D, Delhanty JD. Chromosome abnormalities in the human oocyte. Cytogenet Genome Res 2011;133:107-18. https://doi.org/10.1159/000323801
  56. Hassold T, Hunt P. To err (meiotically) is human: the genesis of human aneuploidy. Nat Rev Genet 2001;2:280-91. https://doi.org/10.1038/35066065
  57. Nagaoka SI, Hassold TJ, Hunt PA. Human aneuploidy: mechanisms and new insights into an age-old problem. Nat Rev Genet 2012;13:493-504. https://doi.org/10.1038/nrg3245
  58. Ghosh S, Hong CS, Feingold E, Ghosh P, Ghosh P, Bhaumik P, et al. Epidemiology of Down syndrome: new insight into the multidimensional interactions among genetic and environmental risk factors in the oocyte. Am J Epidemiol 2011;174:1009-16. https://doi.org/10.1093/aje/kwr240
  59. Zheng CJ, Byers B. Oocyte selection: a new model for the maternal-age dependence of Down syndrome. Hum Genet 1992;90:1-6. https://doi.org/10.1007/BF00210736
  60. Bolcun-Filas E, Rinaldi VD, White ME, Schimenti JC. Reversal of female infertility by Chk2 ablation reveals the oocyte DNA damage checkpoint pathway. Science 2014;343:533-6. https://doi.org/10.1126/science.1247671
  61. Stringer JM, Winship A, Zerafa N, Wakefield M, Hutt K. Oocytes can efficiently repair DNA double-strand breaks to restore genetic integrity and protect offspring health. Proc Natl Acad Sci U S A 2020;117:11513-22. https://doi.org/10.1073/pnas.2001124117
  62. Tsafriri A, Reich R. Molecular aspects of mammalian ovulation. Exp Clin Endocrinol Diabetes 1999;107:1-11. https://doi.org/10.1055/s-0029-1212066
  63. Mastroianni L, Coutifaris C. The FIGO manual of human reproduction. Vol 1. Reproductive physiology. Park Ridge: Parthenon Publishing; 1990.
  64. Espey LL. Ovulation as an inflammatory reaction: a hypothesis. Biol Reprod 1980;22:73-106. https://doi.org/10.1095/biolreprod22.1.73
  65. Duffy DM, Ko C, Jo M, Brannstrom M, Curry TE. Ovulation: parallels with inflammatory processes. Endocr Rev 2019;40:369-416. https://doi.org/10.1210/er.2018-00075
  66. Bukulmez O, Arici A. Leukocytes in ovarian function. Hum Reprod Update 2000;6:1-15. https://doi.org/10.1093/humupd/6.1.1
  67. Fleming JS, Beaugie CR, Haviv I, Chenevix-Trench G, Tan OL. Incessant ovulation, inflammation and epithelial ovarian carcinogenesis: revisiting old hypotheses. Mol Cell Endocrinol 2006;247:4-21. https://doi.org/10.1016/j.mce.2005.09.014
  68. Oakley OR, Kim H, El-Amouri I, Lin PC, Cho J, Bani-Ahmad M, et al. Periovulatory leukocyte infiltration in the rat ovary. Endocrinology 2010;151:4551-9. https://doi.org/10.1210/en.2009-1444
  69. Yamada M, Gentry PA. Hemostatic profile of bovine ovarian follicular fluid. Can J Physiol Pharmacol 1995;73:624-9. https://doi.org/10.1139/y95-079
  70. Shukovski L, Tsafriri A. The involvement of nitric oxide in the ovulatory process in the rat. Endocrinology 1994;135:2287-90. https://doi.org/10.1210/en.135.5.2287
  71. Yamauchi J, Miyazaki T, Iwasaki S, Kishi I, Kuroshima M, Tei C, et al. Effects of nitric oxide on ovulation and ovarian steroidogenesis and prostaglandin production in the rabbit. Endocrinology 1997;138:3630-7. https://doi.org/10.1210/en.138.9.3630
  72. Jablonka-Shariff A, Olson LM. The role of nitric oxide in oocyte meiotic maturation and ovulation: meiotic abnormalities of endothelial nitric oxide synthase knock-out mouse oocytes. Endocrinology 1998;139:2944-54. https://doi.org/10.1210/en.139.6.2944
  73. Ness RB, Cottreau C. Possible role of ovarian epithelial inflammation in ovarian cancer. J Natl Cancer Inst 1999;91:1459-67. https://doi.org/10.1093/jnci/91.17.1459
  74. Kodaman PH, Behrman HR. Endocrine-regulated and protein kinase C-dependent generation of superoxide by rat preovulatory follicles. Endocrinology 2001;142:687-93. https://doi.org/10.1210/en.142.2.687
  75. Park CJ, Lin PC, Zhou S, Barakat R, Bashir ST, Choi JM, et al. Progesterone receptor serves the ovary as a trigger of ovulation and a terminator of inflammation. Cell Rep 2020;31:107496. https://doi.org/10.1016/j.celrep.2020.03.060
  76. Gerard N, Caillaud M, Martoriati A, Goudet G, Lalmanach AC. The interleukin-1 system and female reproduction. J Endocrinol 2004;180:203-12. https://doi.org/10.1677/joe.0.1800203
  77. Needleman P, Turk J, Jakschik BA, Morrison AR, Lefkowith JB. Arachidonic acid metabolism. Annu Rev Biochem 1986;55:69-102. https://doi.org/10.1146/annurev.bi.55.070186.000441
  78. DeWitt DL. Prostaglandin endoperoxide synthase: regulation of enzyme expression. Biochim Biophys Acta 1991;1083:121-34. https://doi.org/10.1016/0005-2760(91)90032-D
  79. Jabbour HN, Sales KJ, Catalano RD, Norman JE. Inflammatory pathways in female reproductive health and disease. Reproduction 2009;138:903-19. https://doi.org/10.1530/REP-09-0247
  80. Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol 2011;31:986-1000. https://doi.org/10.1161/ATVBAHA.110.207449
  81. Murdoch WJ, Townsend RS, McDonnel AC. Ovulation-induced DNA damage in ovarian surface epithelial cells of ewes: prospective regulatory mechanisms of repair/survival and apoptosis. Biol Reprod 2001;65:1417-24. https://doi.org/10.1095/biolreprod65.5.1417
  82. Savant SS, Sriramkumar S, O'Hagan HM. The role of inflammation and inflammatory mediators in the development, progression, metastasis, and chemoresistance of epithelial ovarian cancer. Cancers (Basel) 2018;10:251. https://doi.org/10.3390/cancers10080251
  83. Jia D, Nagaoka Y, Katsumata M, Orsulic S. Inflammation is a key contributor to ovarian cancer cell seeding. Sci Rep 2018;8:12394. https://doi.org/10.1038/s41598-018-30261-8
  84. Yang-Hartwich Y, Gurrea-Soteras M, Sumi N, Joo WD, Holmberg JC, Craveiro V, et al. Ovulation and extra-ovarian origin of ovarian cancer. Sci Rep 2014;4:6116. https://doi.org/10.1038/srep06116
  85. Spanel-Borowski K. Ovulation as danger signaling event of innate immunity. Mol Cell Endocrinol 2011;333:1-7. https://doi.org/10.1016/j.mce.2010.12.008
  86. Lliberos C, Liew SH, Zareie P, La Gruta NL, Mansell A, Hutt K. Evaluation of inflammation and follicle depletion during ovarian ageing in mice. Sci Rep 2021;11:278. https://doi.org/10.1038/s41598-020-79488-4
  87. Huang Y, Hu C, Ye H, Luo R, Fu X, Li X, et al. Inflamm-aging: a new mechanism affecting premature ovarian insufficiency. J Immunol Res 2019;2019:8069898. https://doi.org/10.1155/2019/8069898
  88. Navarro-Pando JM, Alcocer-Gomez E, Castejon-Vega B, Navarro-Villaran E, Condes-Hervas M, Mundi-Roldan M, et al. Inhibition of the NLRP3 inflammasome prevents ovarian aging. Sci Adv 2021;7:eabc7409. https://doi.org/10.1126/sciadv.abc7409
  89. Yang L, Chen Y, Liu Y, Xing Y, Miao C, Zhao Y, et al. The role of oxidative stress and natural antioxidants in ovarian aging. Front Pharmacol 2021;11:617843. https://doi.org/10.3389/fphar.2020.617843
  90. Sammaritano LR. Menopause in patients with autoimmune diseases. Autoimmun Rev 2012;11:A430-6. https://doi.org/10.1016/j.autrev.2011.11.006
  91. Reato G, Morlin L, Chen S, Furmaniak J, Smith BR, Masiero S, et al. Premature ovarian failure in patients with autoimmune Addison's disease: clinical, genetic, and immunological evaluation. J Clin Endocrinol Metab 2011;96:E1255-61. https://doi.org/10.1210/jc.2011-0414
  92. Coulam CB, Stern JJ. Immunology of ovarian failure. Am J Reprod Immunol 1991;25:169-74. https://doi.org/10.1111/j.1600-0897.1991.tb01089.x
  93. Naz RK, Thurston D, Santoro N. Circulating tumor necrosis factor (TNF)-alpha in normally cycling women and patients with premature ovarian failure and polycystic ovaries. Am J Reprod Immunol 1995;34:170-5. https://doi.org/10.1111/j.1600-0897.1995.tb00934.x
  94. Rybinski B, Franco-Barraza J, Cukierman E. The wound healing, chronic fibrosis, and cancer progression triad. Physiol Genomics 2014;46:223-44. https://doi.org/10.1152/physiolgenomics.00158.2013
  95. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 2012;18:1028-40. https://doi.org/10.1038/nm.2807
  96. Mellembakken JR, Berga SL, Kilen M, Tanbo TG, Abyholm T, Fedorcsak P. Sustained fertility from 22 to 41 years of age in women with polycystic ovarian syndrome. Hum Reprod 2011;26:2499-504. https://doi.org/10.1093/humrep/der214
  97. Webber LJ, Stubbs SA, Stark J, Margara RA, Trew GH, Lavery SA, et al. Prolonged survival in culture of preantral follicles from polycystic ovaries. J Clin Endocrinol Metab 2007;92:1975-8. https://doi.org/10.1210/jc.2006-1422
  98. Wiser A, Shalom-Paz E, Hyman JH, Sokal-Arnon T, Bantan N, Holzer H, et al. Age-related normogram for antral follicle count in women with polycystic ovary syndrome. Reprod Biomed Online 2013;27:414-8. https://doi.org/10.1016/j.rbmo.2013.06.016
  99. Iversen L, Fielding S, Lidegaard O, Morch LS, Skovlund CW, Hannaford PC. Association between contemporary hormonal contraception and ovarian cancer in women of reproductive age in Denmark: prospective, nationwide cohort study. BMJ 2018;362:k3609.
  100. Auphan N, DiDonato JA, Rosette C, Helmberg A, Karin M. Immunosuppression by glucocorticoids: inhibition of NF-kappa B activity through induction of I kappa B synthesis. Science 1995;270:286-90. https://doi.org/10.1126/science.270.5234.286
  101. Barkan D, Jia H, Dantes A, Vardimon L, Amsterdam A, Rubinstein M. Leptin modulates the glucocorticoid-induced ovarian steroidogenesis. Endocrinology 1999;140:1731-8. https://doi.org/10.1210/en.140.4.1731