Skip to main content

Advertisement

Log in

Aberrant H19 Expression Disrupts Ovarian Cyp17 and Testosterone Production and Is Associated with Polycystic Ovary Syndrome in Women

  • Reproductive Endocrinology: Original Article
  • Published:
Reproductive Sciences Aims and scope Submit manuscript

Abstract

As one of the most common endocrine disorders affecting women, polycystic ovary syndrome (PCOS) is associated with serious conditions including anovulation, endometrial cancer, infertility, hyperandrogenemia, and an increased risk for obesity and metabolic derangements. One contributing etiology to the pathophysiology of hyperandrogenemia associated with PCOS is an intrinsic alteration in ovarian steroidogenesis, leading to enhanced synthesis of androgens including testosterone. Studies have suggested that the increased testosterone synthesis seen in PCOS is driven in part by increased activity of CYP17A1, the rate-limiting enzyme for the formation of androgens in the gonads and adrenal cortex, which represents a critical factor driving enhanced testosterone secretion in PCOS. In this work, we evaluated the hypothesis that dysregulation of the noncoding RNA H19 results in aberrant CYP17 and testosterone production. To achieve this, we measured Cyp17 in ovarian tissues of H19 knockout mice, and quantified serum testosterone levels, in comparison with wild-type controls. We also evaluated circulating and ovarian H19 expression and correlated results with the presence or absence of PCOS in a group of women undergoing evaluation and treatment for infertility. We found that the loss of H19 in a mouse model results in decreased ovarian Cyp17, along with decreased serum testosterone in female mice. Moreover, utilizing serum samples and cumulus cells from women with PCOS, we showed that circulating and ovarian levels of H19 are increased in women with PCOS compared to controls. Findings from our multimodal experimental strategy, involving both a mouse model of dysregulated H19 expression and clinical serum and ovarian cellular samples from women with PCOS, suggest that the loss of H19 may disrupt androgen production via a Cyp17-mediated mechanism. Conversely, excess H19 may play a role in the pathogenesis of PCOS-associated hyperandrogenemia.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Data Availability

The raw data used to support the findings of this study are available upon request.

References

  1. Bozdag G, Mumusoglu S, Zengin D, Karabulut E, Yildiz BO. The prevalence and phenotypic features of polycystic ovary syndrome: a systematic review and meta-analysis. Hum Reprod. 2016;31(12):2841–55. https://doi.org/10.1093/humrep/dew218.

    Article  PubMed  Google Scholar 

  2. Randeva HS, Tan BK, Weickert MO, et al. Cardiometabolic aspects of the polycystic ovary syndrome. Endocr Rev. 2012;33(5):812–41. https://doi.org/10.1210/er.2012-1003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gottschau M, Kjaer SK, Jensen A, Munk C, Mellemkjaer L. Risk of cancer among women with polycystic ovary syndrome: a Danish cohort study. Gynecol Oncol. 2015;136(1):99–103. https://doi.org/10.1016/j.ygyno.2014.11.012.

    Article  PubMed  Google Scholar 

  4. Azziz R, Woods KS, Reyna R, Key TJ, Knochenhauer ES, Yildiz BO. The prevalence and features of the polycystic ovary syndrome in an unselected population. J Clin Endocrinol Metab. 2004;89(6):2745–9. https://doi.org/10.1210/jc.2003-032046.

    Article  CAS  PubMed  Google Scholar 

  5. Lerchbaum E, Schwetz V, Rabe T, Giuliani A, Obermayer-Pietsch B. Hyperandrogenemia in polycystic ovary syndrome: exploration of the role of free testosterone and androstenedione in metabolic phenotype. PLoS ONE. 2014;9(10):e108263–e108263. https://doi.org/10.1371/journal.pone.0108263.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Rosenfield RL, Ehrmann DA. The pathogenesis of polycystic ovary syndrome (PCOS): the hypothesis of PCOS as functional ovarian hyperandrogenism revisited. Endocr Rev. 2016;37(5):467–520. https://doi.org/10.1210/er.2015-1104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Nelson VL, Qin KN, Rosenfield RL, et al. The biochemical basis for increased testosterone production in theca cells propagated from patients with polycystic ovary syndrome. J Clin Endocrinol Metab. 2001;86(12):5925–33. https://doi.org/10.1210/jcem.86.12.8088.

    Article  CAS  PubMed  Google Scholar 

  8. Wickenheisser JK, Quinn PG, Nelson VL, Legro RS, Strauss JF 3rd, McAllister JM. Differential activity of the cytochrome P450 17alpha-hydroxylase and steroidogenic acute regulatory protein gene promoters in normal and polycystic ovary syndrome theca cells. J Clin Endocrinol Metab. 2000;85(6):2304–11. https://doi.org/10.1210/jcem.85.6.6631.

    Article  CAS  PubMed  Google Scholar 

  9. Cech TR, Steitz JA. Review The noncoding RNA revolution — trashing old rules to forge new ones. Cell. 2014;157(1):77–94. https://doi.org/10.1016/j.cell.2014.03.008.

    Article  CAS  PubMed  Google Scholar 

  10. Yao R-W, Wang Y, Chen L-L. Cellular functions of long noncoding RNAs. Nat Cell Biol. 2019;21(5):542–51. https://doi.org/10.1038/s41556-019-0311-8.

    Article  CAS  PubMed  Google Scholar 

  11. Zhang R, Wesevich V, Chen Z, Zhang D, Kallen AN. Emerging roles for noncoding RNAs in female sex steroids and reproductive disease. Mol Cell Endocrinol. 2020;518:110875. https://doi.org/10.1016/j.mce.2020.110875.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Men Y, Fan Y, Shen Y, Lu L, Kallen AN. The steroidogenic acute regulatory protein (StAR) is regulated by the H19/let-7 axis. Endocrinology. 2017;158(2):402–9. https://doi.org/10.1210/en.2016-1340.

    Article  CAS  PubMed  Google Scholar 

  13. Gabory A, Ripoche M-A, Le Digarcher A, et al. H19 acts as a trans regulator of the imprinted gene network controlling growth in mice. Development. 2009;136(20):3413–21. https://doi.org/10.1242/dev.036061.

    Article  CAS  PubMed  Google Scholar 

  14. Ripoche MA, Kress C, Poirier F, Dandolo L. Deletion of the H19 transcription unit reveals the existence of a putative imprinting control element. Genes Dev. 1997;11(12):1596–604.

    Article  CAS  Google Scholar 

  15. Men Y, Fan Y, Shen Y, Lu L, Kallen AN. The steroidogenic acute regulatory protein (StAR) is regulated by the H19/let-7 axis. Endocrinology. 2017;158(2):402–9.

    Article  CAS  Google Scholar 

  16. Caligioni CS. Assessing reproductive status/stages in mice. Curr Protoc Neurosci. 2009;(SUPPL. 48):1–11. https://doi.org/10.1002/0471142301.nsa04is48

  17. Wang J, Zhao H, Fan Z, et al. Long noncoding RNA H19 promotes neuroinflammation in ischemic stroke by driving histone deacetylase 1–dependent M1 microglial polarization. Stroke. 2017;48(8):2211–21. https://doi.org/10.1161/STROKEAHA.117.017387.

    Article  CAS  PubMed  Google Scholar 

  18. Wu Y, Wang Y-Q, Weng W-W, et al. A serum-circulating long noncoding RNA signature can discriminate between patients with clear cell renal cell carcinoma and healthy controls. Oncogenesis. 2016;5(2): e192. https://doi.org/10.1038/oncsis.2015.48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wang J, Yang K, Yuan W, Gao Z. Determination of serum exosomal H19 as a noninvasive biomarker for bladder cancer diagnosis and prognosis. Med Sci Monit. 2018;24:9307–16. https://doi.org/10.12659/msm.912018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Qin C, Xia X, Fan Y, et al. A novel, noncoding-RNA-mediated, post-transcriptional mechanism of anti-Mullerian hormone regulation by the H19/let-7 axis. Biol Reprod. 2018;0(August):1–11.

  21. Moller DE, Flier JS. Insulin resistance — mechanisms, syndromes, and implications. N Engl J Med. 1991;325(13):938–48. https://doi.org/10.1056/NEJM199109263251307.

    Article  CAS  PubMed  Google Scholar 

  22. Lorenzo C, Haffner SM, Stančáková A, Laakso M. Relation of direct and surrogate measures of insulin resistance to cardiovascular risk factors in nondiabetic finnish offspring of type 2 diabetic individuals. J Clin Endocrinol Metab. 2010;95(11):5082–90. https://doi.org/10.1210/jc.2010-1144.

    Article  CAS  PubMed  Google Scholar 

  23. Su AI, Cooke MP, Ching KA, et al. Large-scale analysis of the human and mouse transcriptomes. Proc Natl Acad Sci. 2002;99(7):4465–70.

    Article  CAS  Google Scholar 

  24. Aherrahrou R, Kulle AE, Alenina N, et al. CYP17A1 deficient XY mice display susceptibility to atherosclerosis, altered lipidomic profile and atypical sex development. Sci Rep. 2020;10(1):1–11. https://doi.org/10.1038/s41598-020-65601-0.

    Article  CAS  Google Scholar 

  25. Ashraf S, Rasool SUA, Nabi M, et al. CYP17 gene polymorphic sequence variation is associated with hyperandrogenism in Kashmiri women with polycystic ovarian syndrome. Gynecol Endocrinol Off J Int Soc Gynecol Endocrinol. 2021;37(3):230–4. https://doi.org/10.1080/09513590.2020.1770724.

    Article  CAS  Google Scholar 

  26. Rahimi Z, Mohammadi M Sc E. The CYP17 MSP AI (T-34C) and CYP19A1 (Trp39Arg) variants in polycystic ovary syndrome: a case-control study. Int J Reprod Biomed. 2019;17(3):201–8. https://doi.org/10.18502/ijrm.v17i3.4519.

    Article  CAS  PubMed Central  Google Scholar 

  27. Panda PK, Rane R, Ravichandran R, Singh S, Panchal H. Genetics of PCOS: A systematic bioinformatics approach to unveil the proteins responsible for PCOS. Genomics data. 2016;8:52–60. https://doi.org/10.1016/j.gdata.2016.03.008.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Li Y, Liu F, Luo S, Hu H, Li X-H, Li S-W. Polymorphism T→C of gene CYP17 promoter and polycystic ovary syndrome risk: A meta-analysis. Gene. 2012;495(1):16–22. https://doi.org/10.1016/j.gene.2011.12.048.

    Article  CAS  PubMed  Google Scholar 

  29. Pusalkar M, Meherji P, Gokral J, Chinnaraj S, Maitra A. CYP11A1 and CYP17 promoter polymorphisms associate with hyperandrogenemia in polycystic ovary syndrome. Fertil Steril. 2009;92(2):653–9. https://doi.org/10.1016/j.fertnstert.2008.07.016.

    Article  CAS  PubMed  Google Scholar 

  30. Munawar Lone N, Babar S, Sultan S, Malik S, Nazeer K, Riaz S. Association of the CYP17 and CYP19 gene polymorphisms in women with polycystic ovary syndrome from Punjab, Pakistan. Gynecol Endocrinol Off J Int Soc Gynecol Endocrinol. September 2020:1–6. https://doi.org/10.1080/09513590.2020.1822803

  31. Li Y, Liang X, Wei L, et al. Study of RNA interference inhibiting rat ovarian androgen biosynthesis by depressing 17alpha-hydroxylase/17, 20-lyase activity in vivo. Reprod Biol Endocrinol. 2009;7:73. https://doi.org/10.1186/1477-7827-7-73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Guttman M, Rinn JL. Modular regulatory principles of large non-coding RNAs. Nature. 2012;482(345):1240–1. https://doi.org/10.1038/nature10887.

    Article  CAS  Google Scholar 

  33. Schorderet P, Duboule D. Structural and functional differences in the long non-coding RNA Hotair in mouse and human. PLoS Genet. 2011;7(5):1–10. https://doi.org/10.1371/journal.pgen.1002071.

    Article  CAS  Google Scholar 

  34. Kallen AN, Zhou XB, Xu J, et al. The imprinted H19 LncRNA antagonizes Let-7 microRNAs. Molefile///Users/amandakallen/Downloads/Gabory_et_al-2010-BioEssays.pdfcular Cell. 2013;52(1):101–112.

  35. Ørom UA, Shiekhattar R. Minireview long noncoding RNAs usher in a new era in the biology of enhancers. Cell. 2013;154(6):1190–3. https://doi.org/10.1016/j.cell.2013.08.028.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Rachmilewitz J, Goshen R, Ariel I, Schneider T, de Groot N, Hochberg A. Parental imprinting of the human H19 gene. FEBS Lett. 1992;309(1):25–8. https://doi.org/10.1016/0014-5793(92)80731-U.

    Article  CAS  PubMed  Google Scholar 

  37. Ariel I, Weinstein D, Voutilainen R, et al. The expression of the imprinted gene H19 in the human female reproductive organs. Diagnostic Mol Pathol. 1997;61(1):17–25.

    Article  Google Scholar 

  38. Khatib H, Schutzkus V. The expression profile of the H19 gene in cattle. Mamm Genome. 2006;17(9):991–6. https://doi.org/10.1007/s00335-006-0038-2.

    Article  CAS  PubMed  Google Scholar 

  39. Dai A, Sun H, Fang T, et al. MicroRNA-133b stimulates ovarian estradiol synthesis by targeting Foxl2. FEBS Lett. 2013;587(15):2474–82. https://doi.org/10.1016/j.febslet.2013.06.023.

    Article  CAS  PubMed  Google Scholar 

  40. Chen B, Xu P, Wang J, Zhang C. The role of MiRNA in polycystic ovary syndrome (PCOS). Gene. 2019;706(March):91–6. https://doi.org/10.1016/j.gene.2019.04.082.

    Article  CAS  PubMed  Google Scholar 

  41. Fu L lu, Xu Y, Li D dan, et al. Expression profiles of mRNA and long noncoding RNA in the ovaries of letrozole-induced polycystic ovary syndrome rat model through deep sequencing. Gene. 2018;657(February):19–29. https://doi.org/10.1016/j.gene.2018.03.002

  42. Huang X, Hao C, Bao H, Wang M, Dai H. Aberrant expression of long noncoding RNAs in cumulus cells isolated from PCOS patients. J Assist Reprod Genet. 2016;33(1):111–21. https://doi.org/10.1007/s10815-015-0630-z.

    Article  PubMed  Google Scholar 

  43. Zhu H, Shyh-Chang N, Segrè AV, et al. The Lin28/let-7 axis regulates glucose metabolism. Cell. 2011;147(1):81–94. https://doi.org/10.1016/j.cell.2011.08.033.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Frost RJA, Olson EN. Control of glucose homeostasis and insulin sensitivity by the Let-7 family of microRNAs. Proc Natl Acad Sci U S A. 2011;108(52):21075–80. https://doi.org/10.1073/pnas.1118922109.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Barbieri RL, Makris A, Randall RW, Daniels G, Kistner RW, Ryan KJ. Insulin stimulates androgen accumulation in incubations of ovarian stroma obtained from women with hyperandrogenism. J Clin Endocrinol Metab. 1986;62(5):904–10. https://doi.org/10.1210/jcem-62-5-904.

    Article  CAS  PubMed  Google Scholar 

  46. Qin L, Huang C, Yan X, Wang Y, Li Z, Wei X. Long non-coding RNA H19 is associated with polycystic ovary syndrome in Chinese women: a preliminary study. Endocr J. 2019. https://doi.org/10.1507/endocrj.ej19-0004.

    Article  PubMed  Google Scholar 

  47. Ou L, Wang D, Zhang H, Yu Q, Hua F. Decreased expression of MiR-138-5p by LncRNA H19 in cervical cancer promotes tumor proliferation. Oncol Res Featur Preclin Clin Cancer Ther. 2017. https://doi.org/10.3727/096504017X15017209042610.

    Article  Google Scholar 

  48. Zhou W, Ye X-L, Xu J, et al. The lncRNA H19 mediates breast cancer cell plasticity during EMT and MET plasticity by differentially sponging miR-200b/c and let-7b. Sci Signal. 2017;10(483). https://doi.org/10.1126/scisignal.aak9557

  49. Li Z, Li Y, Li Y, et al. Long non-coding RNA H19 promotes the proliferation and invasion of breast cancer through upregulating DNMT1 expression by sponging miR-152. J Biochem Mol Toxicol. 2017;31(e21933):1–9. https://doi.org/10.1002/jbt.21933.

    Article  CAS  PubMed  Google Scholar 

  50. Imig J, Brunschweiger A, Brümmer A, et al. miR-CLIP capture of a miRNA targetome uncovers a lincRNA H19–miR-106a interaction. Nat Chem Biol. 2014;11(2):107–14. https://doi.org/10.1038/nchembio.1713.

    Article  CAS  PubMed  Google Scholar 

  51. Dumesic DA, Oberfield SE, Stener-Victorin E, Marshall JC, Laven JS, Legro RS. Scientific statement on the diagnostic criteria, epidemiology, pathophysiology, and molecular genetics of polycystic ovary syndrome. Endocr Rev. 2015;36(5):487–525. https://doi.org/10.1210/er.2015-1018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wang Q, Shang J, Zhang Y, Zhou W. Metformin and sitagliptin combination therapy ameliorates polycystic ovary syndrome with insulin resistance through upregulation of lncRNA H19. Cell Cycle. 2019;18(19):2538–49. https://doi.org/10.1080/15384101.2019.1652036.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Norman RJ, Dewailly D, Legro RS, Hickey TE. Polycystic ovary syndrome. Lancet (London, England). 2007;370(9588):685–97. https://doi.org/10.1016/S0140-6736(07)61345-2.

    Article  CAS  Google Scholar 

  54. Palomba S, Orio F, Falbo A, et al. Prospective parallel randomized, double-blind, double-dummy controlled clinical trial comparing clomiphene citrate and metformin as the first-line treatment for ovulation induction in nonobese anovulatory women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2005;90(7):4068–74. https://doi.org/10.1210/jc.2005-0110.

    Article  CAS  PubMed  Google Scholar 

  55. Mansfield R, Galea R, Brincat M, Hole D, Mason H. Metformin has direct effects on human ovarian steroidogenesis. Fertil Steril. 2003;79(4):956–62. https://doi.org/10.1016/S0015-0282(02)04925-7.

    Article  PubMed  Google Scholar 

  56. Attia GR, Rainey WE, Carr BR. Metformin directly inhibits androgen production in human thecal cells. Fertil Steril. 2001;76(3):517–24. https://doi.org/10.1016/S0015-0282(01)01975-6.

    Article  CAS  PubMed  Google Scholar 

  57. Zhong T, Men Y, Lu L, et al. Metformin alters DNA methylation genome-wide via the H19/SAHH axis. Oncogene. 2017;36(17):2345–54. https://doi.org/10.1038/onc.2016.391.

    Article  CAS  PubMed  Google Scholar 

  58. La Marca A, Morgante G, Paglia T, Ciotta L, Cianci A, De Leo V. Effects of metformin on adrenal steroidogenesis in women with polycystic ovary syndrome. Fertil Steril. 1999;72(6):985–9. https://doi.org/10.1016/S0015-0282(99)00407-0.

    Article  PubMed  Google Scholar 

  59. Chen Z, Wei H, Zhao X, et al. Metformin treatment alleviates polycystic ovary syndrome by decreasing the expression of MMP-2 and MMP-9 via H19/miR-29b-3p and AKT/mTOR/autophagy signaling pathways. J Cell Physiol. 2019;234(11):19964–76. https://doi.org/10.1002/jcp.28594.

    Article  CAS  PubMed  Google Scholar 

  60. Zhou X, Yin C, Dang Y, Ye F, Zhang G. Identification of the long non-coding RNA H19 in plasma as a novel biomarker for diagnosis of gastric cancer. Sci Rep. 2015;5(May):1–10. https://doi.org/10.1038/srep11516.

    Article  Google Scholar 

  61. Hashad D, Elbanna A, Ibrahim A, Khedr G. Evaluation of the role of circulating long non-coding RNA H19 as a promising novel biomarker in plasma of patients with gastric cancer. J Clin Lab Anal. 2016;30(6):1100–5.

    Article  CAS  Google Scholar 

  62. Xia X, Burn M, Chen Y, Johnson J, Kallen A. The relationship between H19 and parameters of ovarian reserve. Reprod Biol Endocrinol. 2020;In press.

  63. Dapas M, Lin FTJ, Nadkarni GN, et al. Distinct subtypes of polycystic ovary syndrome with novel genetic associations: an unsupervised, phenotypic clustering analysis. PLoS Med. 2020;17(6):e1003132–e1003132. https://doi.org/10.1371/journal.pmed.1003132.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sakian S, Louie K, Wong EC, et al. Altered gene expression of H19 and IGF2 in placentas from ART pregnancies. Placenta. 2015;36(10):1100–5.

    Article  CAS  Google Scholar 

  65. Li T, Vu TH, Ulaner GA, et al. IVF results in de novo DNA methylation and histone methylation at an Igf2-H19 imprinting epigenetic switch. Mol Hum Reprod. 2005;11(9):631–40.

    Article  CAS  Google Scholar 

  66. Shi X, Ni Y, Zheng H, et al. Abnormal methylation patterns at the IGF2/H19 imprinting control region in phenotypically normal babies conceived by assisted reproductive technologies. Eur J Obstet Gynecol Reprod Biol. 2011;158(1):52–5.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

H19KO mice were provided by Luisa Dandolo, PhD, and Stefan Muljo, PhD. The University of Virginia Center for Research in Reproduction Ligand Assay and Analysis Core (used for serum steroid hormone analysis) is supported by the Eunice Kennedy Shriver NICHD Grant R24 HD102061. We thank Ms. Meirav Sela for her assistance with manuscript proofreading.

Funding

Dr Kallen received funding and research support provided by the NIH-NICHD (R01HD101475), the Reproductive Scientist Development Program (NIH-NICHD Project #2K12HD000849-26), the American Society for Reproductive Medicine, and the NIH Loan Repayment Program. Dr Kallen and Dr Xi received funding and support from the Milstein Medical Asian American partnership Foundation (MMAAPF). The University of Virginia Center for Research in Reproduction Ligand Assay and Analysis Core (used for serum steroid hormone analysis) is supported by the Eunice Kennedy Shriver NICHD Grant R24 HD102061.

Author information

Authors and Affiliations

Authors

Contributions

AK, XX, and LL conceived and planned the experiments. XX, LL, ZC, and CK carried out the experiments. XX and CK contributed to sample collection and preparation. XX, ZC, LL, and MB contributed to the interpretation of the results. AK and ZC took the lead in writing the manuscript. All authors provided critical feedback and helped shape the research, analysis, and manuscript.

Corresponding author

Correspondence to Amanda N. Kallen.

Ethics declarations

Ethics Approvals

Studies involving mice were approved by the Yale University Institutional Animal Care and Use Committee (IACUC protocol #2021–20018). Studies utilizing patient samples were approved by the Gazi University Institutional Review Board committee (IRB protocol #131/11.05.2011) and the Yale University Institutional Review Board (IRB protocol #1606017946).

Conflict of Interest

The authors declare no competing interests.

Additional information

Zhaojuan Chen and Lan Liu are co-first authors.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Z., Liu, L., Xi, X. et al. Aberrant H19 Expression Disrupts Ovarian Cyp17 and Testosterone Production and Is Associated with Polycystic Ovary Syndrome in Women. Reprod. Sci. 29, 1357–1367 (2022). https://doi.org/10.1007/s43032-021-00700-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s43032-021-00700-5

Keywords

Navigation