Skip to main navigation menu Skip to main content Skip to site footer

Original Article

Vol. 6 No. 1

Endocrine activities modulated by adipose-mesenchymal stem cell in an animal model induced to polycystic ovary syndrome

Published
2024-04-17

Abstract

Purpose - Animal models offer a useful way to study the reproductive and metabolic abnormalities, including PCOS. MSCs have received increasing attention as a potential cell-based therapy and regenerative medicine, due to their effects in modulation of different molecular and biological pathways. The aims of the present work were to investigate modulation of the ovarian microenvironment by AdMSCs in an animal model induced to PCOS. Methods - Female rats were divided into control, polycystic ovary, and mesenchymal stem cell groups, evaluated at two different times after PCOS induction and injection of AdMSCs. Results - The polycystic ovary group showed changes in ovarian cycles, the presence of cysts in the ovaries, and hyperandrogenemia. In addition, changes in plasma insulin, glucose, leptin, and osteocalcin were observed in the polycystic ovary group. These metabolic changes were modulated by the injection of AdMSCs into the ovary. Data are presented for female rats in an animal model integrating PCOS with AdMSCs, together with the relationships among ovaries, bones, and adipocytes. Conclusion - The results suggested the existence of endocrine-metabolic-reproductive microenvironment relationships modulated by AdMSCs, which should help in guiding further investigations to clarify pathophysiological mechanisms that have not yet been fully elucidated.

References

  1. Aflatounian A, Eduwards MC, Paris VR, Bertoldo MJ, Desai R, Glichrist RB. Ledger WL, Handelsman DJ, Walters KA. Androgen signaling pathways driving reproductive and metabolic phenotypes in a PCOS mouse model. J Endocrinol. 2020;245(3):381-395.
  2. Padmanabhan V, Veiga-Lopez A. Sheep models of polycystic ovary syndrome phenotype. Mol Cell Endocrinol. 2013;373:8-20.
  3. Doretto L, Mari FC, Chaves AC. Polycystic ovary syndrome and psychotic disorders. Front Psychiatry. 2020;11:543.
  4. Spallanzani RG. Visceral adipose tissue mesenchymal stroma cells in the intersection of immunology and metabolism. Am J Physiol Endocrinol Metab. 2021;320:E512-E519.
  5. Esfandyari S, Chugh RM, Park H, Hobeika E, Ulin M, Al-Hendy A. Mesenchymal stem cells as a Bio-organ for treatment of female infertility. Cells. 2020;9:2253.
  6. Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential. Trends Pharmacol Sci. 2020;41(9):653-664.
  7. Gomez-Salazar M, Gonzales-Galofre ZN, Casamitijana J, Crisan M, James AW, Péault B. 2020 Five decades later, are mesenchymal stem cells still relevant? Front Bioeng Biotechnol. 2003;8:148.
  8. Maacha S, Sidahmed H, Jacob S, Gentilcore G, Calzone R, Grivel JC, Cugno C. Paracrine mechanisms of mesenchymal stromal cells in angiogenesis. Stem Cell Inter. 2020;9:4356359.
  9. Yang K, Oark HJ, Han S, Lee J, Ko E, Kim J, Lee JS, Yu JH, Song KY, Cheong E, Cho SR, Chung S, Cho SW. Recapitulation of in vivo-like paracrine signals of human mesenchymal stem cells for functional neuronal differentiation of human neural stem cells in a 3D microfluidic system. Biomaterials. 2015;63:177-188.
  10. Segers VF, Lee RT. Stem-cell therapy for cardiac disease. Nature. 2008;451:937-942.
  11. Chen L, Tredget EE, Wu PY, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. Plos One. 2008;3:e1886.
  12. Alves ED, Bonfá ALO, Pigatto GR, Anselmo-Franci JA, Achcar JA, Parizotto NA, Montrezor LH. Photobiomodulation can improve ovarian activity in polycystic ovary syndrome-induced rats. J Photochem Photobiol B Biol. 2019;194:6-13.
  13. Ryu Y, Kim SW, Kim YY, Ku SY. Animal Models for Human Polycystic Ovary Syndrome (PCOS) Focused on the Use of Indirect Hormonal Perturbations: A Review of the Literature. Int J Mol Sci. 2019;20(11):1-20.
  14. Kuang HY, Li Y, Zhang HP, Legro RS, Stener-Victorin E. Data do not support effectiveness of acupuncture for improving live birth rate in women with polycystic ovary syndrome. Chinese J Integ Med. 2018;24(5):399-400.
  15. Caldwell AS, Caldwell ASL, Edwards MC, Desai R, Jimenez H, Gilchrist RB, Handelsman DJ, Walters KA. Neuroendocrine androgen action is a key extraovarian mediator in the development of polycystic ovary syndrome. Proc Natl Acad Sci USA. 2017;114:3334–3343.
  16. Krishnan A, Muthusami S. Hormonal alterations in PCOS and its influence on bone metabolism. J Endocrinol. 2017;232(2):99-113.
  17. Marcondes RR, Maliqueo M, Fornes R, Benrick A, Hu M, Ivarsson N, Carlström M, Cushman SW, Stenkula KG, Maciel GAR, Stener-Victorin E. Exercise differentially affects metabolic functions and white adipose tissue in female letrozole- and dihydrotestosterone-induced mouse models of polycystic ovary syndrome. Mol Cel Endocrinol. 2017;448:66-76.
  18. Walters KA, Allan CM, Handelsman DJ. Rodent models for human polycystic ovary syndrome. Biol Reprod. 2012;86(5):1-12.
  19. Shi D, Vine DF. Animal model of polycystic ovary syndrome: a focused review of rodent models in relationship to clinical phenotypes and cardiometabolic risk. Fert Steril. 2012;98(1):185-193.
  20. Walters KA, Rodriguez PV, Aflatounain A, Handelsman DJ. Androgens and ovarian function: translation from basic discovery research of clinical impact. J Endocrinol. 2019;242(2):R23-R50.
  21. Wickenheisser JK, Nelson-DeGrave VL, McAllister JM. Dysregulation of cytochrome P450 17alpha-hydroxylase messenger ribonucleic acid stability in theca cells isolated from women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2005;90(3):1720-1727.
  22. Paixão L, Ramos RB, Lavarda A, Morsh DM, Spritzer PM. Animal model of hyperandrogenism and ovarian morphology changes as features of polycystic ovary syndrome: a systematic review. Reprod Biol Endocrinol. 2017;15:12.
  23. Stener-Victorin E, Padmanabhan V, Walters KA, Campbell RE, Bernick A, Giacobini P, Dumesic DA, Abbott DH. Animal models to understand the etiology and pathophysiology of polycystic ovary syndrome. Endocr Rev. 2020;41:538-576.
  24. Brawer JR, Munoz M, Farookhi R. Development of the polycystic ovarian condition (PCO) in the estradiol valerate-treated rat. Biol Reprod. 1986;35(3):647-655.
  25. Montrezor LH, Carvalho D, Dias MB, Anselmo-Franci JA, Bícego KC, Gargaglioni LH. Hypoxic and hypercapnic ventilatory responses in rats with polycystic ovaries. Resp Physiol Neurobiol. 2015;217:17–24.
  26. Li C, Li G, Liu M, Zhou T, Zhou H. Paracrine effect of inflammatory cytokine-activated bone marrow mesenchymal stem cell and its role in osteoblast function. J Biosci Bioeng. 2016;121(2):213-219.
  27. Ma Y, Andrisse S, Chen Y, Childress S, Xue P, Wang Z, Jones D, Ko CM, Wu S. Androgen receptor in the ovary theca cells plays a critical role in androgen-induced reproductive dysfunction. Endocrinol. 2017;158(1):98–108.
  28. Caldwell AS, Middleton LJ, Jimenez H, Desai R, Mcmahon AC, Allan CM, Handelsman CJ, Walters KA. Characterization of reproductive, metabolic, and endocrine features of polycystic ovary syndrome in female hyperandrogenic mouse models. Endocrinol. 2014;155:3146-3159.
  29. Van Houten EI, Kramer P, Mcluskey A, Karels B, Themmen AP, Visser JA. Reproductive and metabolic phenotype of a mouse model of PCOS. Endocrinol. 2012;153(6):2861–2869.
  30. Dumesic DA, Padmanabhan V, Chazenbalk GD, Abbott DH. Polycystic ovary syndrome as a plausible evolutionary outcome of metabolic adaptation. Reprod Biol Endocrinol. 2022;20:12.
  31. Oliveira M, Mathias LS, Rodrigues BM, Mariani BG, Graceli JB, De Sibio MT, Olimpio RMC, Moretto FCF, Deprá IC, Nogueira CR. The roles of triiodothyronine and irisin in improving the lipid profile and directing the browning of human adipose subcutaneous cells. Mol Cell Endocrinol. 2020;506:110744.
  32. Amirkhanloo F, Esmaeilzadeh S, Mirabi P, Abedini A, Amiri M, Saghebi R, Golsorkhtabaramiri M. Comparison of Foeniculum Vulgare versus metformin on insulin resistance and anthropometric indices of women with polycystic ovary, an open-label controlled trial study. Obesity Medicine. 2022;31:100401.
  33. Norman RJ, Dewailly D, Legro RS, Hickey TE. Polycystic ovary syndrome. The Lancet. 2007;370:685-597.
  34. Domouky AM, Hegab AS, Al-Shahat A, Raafat N. Mesenchymal stem cells and differentiated insulin producing cell are new horizons for pancreatic regeneration in type I diabetes mellitus. Int J Biochem Cell Biol. 2017;87:77-85.
  35. Wang L, Liu T, Liang R, Wang G, Liu Y, Zou J, Liu N, Zhang B, Liu Y, Ding X, Cai S, Wang Z, Xu X, Ricordi C, Wang S, Shen Z. Mesenchymal stem cells ameliorate ß cell dysfunction of human type 2 diabetic islet by reversing ß cell dedifferentiation. EBioMedicine. 2020;51:102615.
  36. Xie Q, Xiong XL, Xiao N, He K, Chen M, Peng J, Su X, Mei H, Dai Y, Wei D, Lin, G, Cheng L. Mesenchymal stem cells alleviate DHEA-induced polycystic ovary syndrome (PCOS) by inhibiting inflammation in mice. Stem Cells Inter. 2019;9782373.
  37. Abbott DH, Barnett DK, Bruns CM, Dumesic SA. Androgen excess fetal programming of female reproduction: a developmental aetiology for polycystic ovary syndrome? Hum Reprod Update. 2005;11:357-374.
  38. Sullivan SD, Moenter SM. Prenatal androgens alter GABAergic drive to gonadotropin-releasing hormone neurons: implications for a common fertility disorder. Proc Natl Acad Sci USA. 2004;101:7129-7134.
  39. Kawa IA, Masood A, Ganie MA, Fatima Q, Jeelani H, Manzoor S, Rizvi SM, Muzamil M, Rashid F. Bisphenol A (BPA) acts as an endocrine disruptor in women with polycystic ovary syndrome: hormonal and metabolic evaluation. Obesity Medicine. 2019;14:100090.
  40. Malard PF, Peixer MAS, Grazia JG, Brunel HSS, Feres LF, Villarroel CL, Siqueira LGB, Dode MAN, Pogue R, Viana JHM, Carvalho JL. Intraovarian injection of mesenchymal stem cells improve oocyte yield and in vitro embryo production in a bovine model of fertility loss. Scientific Reports. 2020;10:8018.
  41. Dumesic DA, Abbott DH, Eisner JR, Goy RW. Prenatal exposure of female rhesus monkey to testosterone propionate increases serum luteinizing hormone levels in adulthood. Fertil Steril. 1997;67:155-163.
  42. Sharma TP, Herkimer C, West C, Ye W, Birch R, Robinson JE, Foster DL, Padmanabhan V. Fetal programming: prenatal androgen disrupts positive feedback actions of estradiol but does not affect timing of puberty in female sheep. Biol Reprod. 2020;66:924-933.
  43. Wu XY, Li ZL, Wu CY, Liu Y-M, Lin H, Wang S-H, Xiao W-F. Endocrine traits of polycystic ovary syndrome in prenatally androgenized female Sprague-Dawley rats. Endocr J. 2010;57:201-209.
  44. Harrison TNH, Chang RJ. Ovarian response to follicle-stimulating hormone in women with polycystic ovary syndrome is diminished compared to ovulatory controls. Clin Endocrinol. 2020;97:310-318.
  45. Zhao YX, Chen SR, Su PP, Huang FH, Shi YC, Shi QY, Lin S. Using mesenchymal stem cells to treat female infertility: an update on female reproductive diseases. Stem Cells Int. 2019;9071720.
  46. Tan BK, Chen J, Hu J, Amar O, Mattu HS, Adya R, Patel VH, Ramanjaneya M, Lehnert H, Randeva HS. Metformin increases the novel adipokine caronectin/CTRP3 in women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2013;98:E1891-900.
  47. Plati E, Kouskouni E, Malamitsi-Puchner A, Boutsikou M, Kaparos G, Baka S. Visfatin and leptina levels in women with polycystic ovaries undergoing ovarian stimulation. Fertil Steril. 2010;94,:451-1456.
  48. Barber TM, Franks S. Adipocyte biology in polycystic ovary syndrome. Mol Cell Endocrinol. 2013;373:68-76.
  49. Svendsen PF, Christiansen M, Hedley PL, Nilas L, Pedersen SB, Madsbad S. Adipose expression of adipocytokines in women with polycystic ovary syndrome. Fertil Steril. 2012;98:235-41.
  50. Swain JE, Dunn RL, McConnell D, Gonzales-Martinez J, Smith GD. Direct effects of leptin on mouse reproductive function: regulation of follicular, oocyte, and embryo development. Biol Reprod. 2004;71(5):1446-1452.
  51. Ruiz-Cortés ZT, Martel-Kennes Y, Gévry NY, Downey BR, Palin M-F, Murphy BD. Biphasic effects of leptin in porcine granulosa cells. Biol Reprod. 2003;68(3):789-796.
  52. Ferron M, Lacombe J. Regulation of energy metabolism by the skeleton: osteocalcin and beyond. Arch Biochem Bioph. 2014;561:137-146.
  53. Shan C, Yue J, Liu W. Broadening the role of osteocalcin in the hypothalamic-pituitary-gonadal axis. J Edocrinol. 2021;249(2):R43-R51.
  54. Hinoi E, Gao N, Jung DY, Yadav V, Yoshizawa T, Myers Jr MG, Chua Jr SC, Kim JK, Kaestner KH, Karsenty G. The sympathetic tone mediates leptin’s inhibition of insulin secretion by modulating osteocalcin bioactivity. J Cell Biol. 2008;183:1235-1242.

Downloads

Download data is not yet available.

Most read articles by the same author(s)