CYP19A1 Promoters Activity in Human Granulosa Cells: A Comparison between PCOS and Normal Subjects

Document Type : Original Article

Authors

1 Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran

2 Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran

3 Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran

4 Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran

5 Department of Genetics at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran

Abstract

Objective: Estrogen, a female hormone maintaining several critical functions in women's physiology, e.g., folliculogenesis and fertility, is predominantly produced by ovarian granulosa cells where aromatase enzyme converts androgen to estrogen. The principal enzyme responsible for this catalytic reaction is encoded by the CYP19A1 gene, with a long  regulatory region. Abnormalities in this process cause metabolic disorders in women, one of the most common of which 
is polycystic ovary syndrome (PCOS). The main purpose of this research was to determine the effect of the promoters 
on aromatase expression in cells with normal and PCOS characteristics. 
Materials and Methods: In this experimental study, four promoters of the CYP19A1 gene, including PII, I.3, I.4, and PII/ I .3 promoter fragments, were cloned upstream of the luciferase gene and transfected into normal and PCOS granulosa cells. Subsequently, the effect of follicle-stimulating hormone (FSH) on the activity of these regulatory regions was examined in the presence and absence of FSH. Western blotting was used to confirm aromatase expression in all groups. Data analysis was performed using ANOVA and paired sample t test, compared by post-hoc least significant difference (LSD) test. 
Results: Luciferase results confirmed the intense activity of PII promoter in the presence of FSH. Moreover, the study 
demonstrated reduced activity of PII promoter in normal granulosa cells, possibly due to the regulatory region of I.3 next to PII. 
Conclusion: FSH stimulates transcription of aromatase enzyme by affecting PII promoter, a process regulated by the 
inhibitory role of the I.3 region in PII activity in granulosa cells. Given the distinct role of these promoters in normal and 
PCOS granulosa cells, the importance of nuclear factors residing in these regions can be discerned. 

Keywords


  1. Homburg R, Insler V. Ovulation induction in perspective. Hum Reprod Update. 2002; 8(5): 449-462.
  2. Matzuk MM, Lamb DJ. The biology of infertility: research advances and clinical challenges. Nat Med. 2008; 14(11): 1197-1213.
  3. Macer ML, Taylor HS. Endometriosis and infertility: a review of the pathogenesis and treatment of endometriosis-associated infertility. Obstet Gynecol Clin North Am. 2012; 39(4): 535-549.
  4. Baldani DP, Skrgatic L, Ougouag R. Polycystic ovary syndrome: important underrecognised cardiometabolic risk factor in reproductive- age women. Int J Endocrinol. 2015; 2015: 786362.
  5. Pangas SA. Regulation of the ovarian reserve by members of the transforming growth factor beta family. Mol Reprod Dev. 2012; 79(10): 666-679.
  6. Kidder GM, Vanderhyden BC. Bidirectional communication between oocytes and follicle cells: ensuring oocyte developmental competence. Can J Physiol Pharmacol. 2010; 88(4): 399-413.
  7. Stocco C. Aromatase expression in the ovary: hormonal and molecular regulation. Steroids. 2008; 73(5): 473-487.
  8. Qiao J, Feng HL. Extra- and intra-ovarian factors in polycystic ovary syndrome: impact on oocyte maturation and embryo developmental competence. Hum Reprod Update. 2011; 17(1): 17-33.
  9. Simpson ER, Clyne C, Rubin G, Boon WC, Robertson K, Britt K, et al. Aromatase--a brief overview. Annu Rev Physiol. 2002; 64: 93-127.
  10. Stocco C. Tissue physiology and pathology of aromatase. Steroids. 2012; 77(1-2): 27-35.
  11. Strauss L, Rantakari P, Sjögren K, Salminen A, Lauren E, Kallio J, et al. Seminal vesicles and urinary bladder as sites of aromatization of androgens in men, evidenced by a CYP19A1-driven luciferase reporter mouse and human tissue specimens. FASEB J. 2013; 27(4): 1342-1350.
  12. Strauss L, Rantakari P, Sjögren K, Salminen A, Lauren E, Kallio J, et al. Seminal vesicles and urinary bladder as sites of aromatization of androgens in men, evidenced by a CYP19A1-driven luciferase reporter mouse and human tissue specimens. FASEB J. 2013; 27(4): 1342-1350.
  13. Demura M, Reierstad S, Innes JE, Bulun SE. Novel promoter I.8 and promoter usage in the CYP19 (aromatase) gene. Reprod Sci. 2008; 15(10): 1044-1053.
  14. Simpson ER, Mahendroo MS, Means GD, Kilgore MW, Hinshelwood MM, Graham-Lorence S, et al. Aromatase cytochrome P450, the enzyme responsible for estrogen biosynthesis. Endocr Rev. 1994; 15(3): 342-355.
  15. Sebastian S, Bulun SE. A highly complex organization of the regulatory region of the human CYP19 (aromatase) gene revealed by the human genome project. J Clin Endocrinol Metab. 2001; 86(10): 4600-4602.
  16. Conley A, Mapes S, Corbin CJ, Greger D, Walters K, Trant J, et al. A comparative approach to structure-function studies of mammalian aromatases. J Steroid Biochem Mol Biol. 2001; 79(1-5): 289-297.
  17. Bulun SE, Lin Z, Imir G, Amin S, Demura M, Yilmaz B, et al. Regulation of aromatase expression in estrogen-responsive breast and uterine disease: from bench to treatment. Pharmacol Rev. 2005; 57(3): 359-83.
  18. Bréard E, Roussel H, Lindet Y, Mittre H, Leymarie P. Presence of exon I.4 mRNA from CYP19 gene in human granulosa cells. Mol Cell Endocrinol. 1999; 154(1-2): 187-190.
  19. Hosseini E, Mehraein F, Shahhoseini M, Karimian L, Nikmard F, Ashrafi M, et al. Epigenetic alterations of CYP19A1 gene in Cumulus cells and its relevance to infertility in endometriosis. J Assist Reprod Genet. 2016; 33(8): 1105-1113.
  20. Hashemian Z, Afsharian P, Farzaneh P, Eftekhari-Yazdi P, Vakhshiteh F, Daneshvar Amoli A, et al. Establishment and characterization of a PCOS and a normal human granulosa cell line. Cytotechnology. 2020; 72(6): 833–845.
  21. Michael MD, Kilgore MW, Morohashi K, Simpson ER. Ad4BP/SF-1 regulates cyclic AMP-induced transcription from the proximal promoter (PII) of the human aromatase P450 (CYP19) gene in the ovary. J Biol Chem. 1995; 270(22): 13561-13566.
  22. Saitoh M, Yanase T, Morinaga H, Tanabe M, Mu YM, Nishi Y, et al. Tributyltin or triphenyltin inhibits aromatase activity in the human granulosa-like tumor cell line KGN. Biochem Biophys Res Commun. 2001; 289(1): 198-204.
  23. Davuluri RV, Suzuki Y, Sugano S, Plass C, Huang TH. The functional consequences of alternative promoter use in mammalian genomes. Trends Genet. 2008; 24(4): 167-177.
  24. Attar E, Bulun SE. Aromatase and other steroidogenic genes in endometriosis: translational aspects. Hum Reprod Update. 2006; 12(1): 49-56.
  25. Fleming NI, Knower KC, Lazarus KA, Fuller PJ, Simpson ER, Clyne CD. Aromatase is a direct target of FOXL2: C134W in granulosa cell tumors via a single highly conserved binding site in the ovarian specific promoter. PLoS One. 2010; 5(12): e14389.
  26. Bates DL, Chen Y, Kim G, Guo L, Chen L. Crystal structures of multiple GATA zinc fingers bound to DNA reveal new insights into DNA recognition and self-association by GATA. J Mol Biol. 2008; 381(5): 1292-1306.
  27. Jin T, Zhang X, Li H, Goss PE. Characterization of a novel silencer element in the human aromatase gene PII promoter. Breast Cancer Res Treat. 2000; 62(2): 151-159.
  28. Ghosh S, Wu Y, Li R, Hu Y. Jun proteins modulate the ovary-specific promoter of aromatase gene in ovarian granulosa cells via a cAMP-responsive element. Oncogene. 2005; 24(13): 2236-2246.
  29. Wu Y, Ghosh S, Nishi Y, Yanase T, Nawata H, Hu Y. The orphan nuclear receptors NURR1 and NGFI-B modulate aromatase gene expression in ovarian granulosa cells: a possible mechanism for repression of aromatase expression upon luteinizing hormone surge. Endocrinology. 2005; 146(1): 237-246.
  30. Sahmi F, Nicola ES, Zamberlam GO, Gonçalves PD, Vanselow J, Price CA. Factors regulating the bovine, caprine, rat and human ovarian aromatase promoters in a bovine granulosa cell model. Gen Comp Endocrinol. 2014; 200: 10-7.
  31. Enjuanes A, Garcia-Giralt N, Supervía A, Nogués X, Ruiz-Gaspà S, Bustamante M, et al. Functional analysis of the I.3, I.6, pII and I.4 promoters of CYP19 (aromatase) gene in human osteoblasts and their role in vitamin D and dexamethasone stimulation. Eur J Endocrinol. 2005; 153(6): 981-988.
  32. Shozu M, Zhao Y, Bulun SE, Simpson ER. Multiple splicing events involved in regulation of human aromatase expression by a novel promoter, I.6. Endocrinology. 1998; 139(4): 1610-1617.
  33. Odawara H, Iwasaki T, Horiguchi J, Rokutanda N, Hirooka K, Miyazaki W, et al. Activation of aromatase expression by retinoic acid receptor-related orphan receptor (ROR) alpha in breast cancer cells: identification of a novel ROR response element. J Biol Chem. 2009; 284(26): 17711-17719.
  34. Chen S. Aromatase and breast cancer. Front Biosci. 1998; 3: d922-33.
  35. Gérard C, Brown KA. Obesity and breast cancer - Role of estrogens and the molecular underpinnings of aromatase regulation in breast adipose tissue. Mol Cell Endocrinol. 2018; 466: 15-30.
  36. Ratre P, Mishra K, Dubey A, Vyas A, Jain A, Thareja S. aromatase inhibitors for the treatment of breast cancer: a journey from the scratch. Anticancer Agents Med Chem. 2020; 20(17): 1994-2004.