The effects of melatonin and metformin on histological characteristics of the ovary and uterus in letrozole-induced polycystic ovarian syndrome mice: A stereological study

Abstract

Background: Polycystic ovarian syndrome (PCOS) with anovulation, hyperandrogenism, ovarian and uterine histological changes, menstrual irregularities, etc. signs is an infertility type. It seems that melatonin and metformin can improve these abnormalities.


Objective: To evaluate the effects of melatonin and metformin on the ovary and uterus in PCOS-induced mice using stereological methods.


Materials and Methods: Seventy-two adult female BALB/c mice (8-wk-old, 20-30 gr) were randomly divided into control (distilled water, gavage), PCOS (90 μg/kg letrozole, gavage), PCOS+metformin (500 mg/kg, gavage), PCOS+melatonin (10 mg/kg, intraperitoneal injection), and PCOS+melatonin control (0.5% ethanol saline) groups (n = 12/each). Another PCOS group was kept for a month to ensure that PCOS features remained. Finally, a stereological evaluation of the uterus and ovary was carried out, and vaginal cytology and serum testosterone levels were assessed.


Results: PCOS mice treated with metformin and melatonin had lower testosterone levels, body weight, and more regular estrus cycles than the PCOS group (p ≤ 0.001). A significant decrease in conglomerate and daughter gland numbers, and primary, secondary, atretic, and cystic follicles numbers with a significant increase in primordial and Graafian follicles, and corpus luteum numbers (p ≤ 0.001) was seen in these treated mice. Also, endometrial vessels’ volume and length significantly increased, but ovarian, endometrial, myometrial, stromal, and glands volume, and endometrial and myometrial thickness dramatically declined (p ≤ 0.001).


Conclusion: It appears that metformin and melatonin could restore the PCOS phenotype including estrus cycle irregularity, high testosterone level, and ovarian and uterine micromorphology to the control levels. However, the 2 treatments had similar effects on the examined parameters.


Key words: Polycystic ovarian syndrome, Melatonin, Metformin, Ovary, Uterus, Mice, Stereology.

References
[1] Deswal R, Narwal V, Dang A, Pundir CS. The prevalence of polycystic ovary syndrome: A brief systematic review. J Hum Reprod Sci 2020; 13: 261–271.

[2] Ayyadurai Th, Thiruppathi SK, Shanmugam A, Diana RKB. Effect of aqueous seed extract of Caesalpinia bonduc (L.) Roxb., on hormonal assay and lipid profile in induced polycystic ovary syndrome albino female rats. Int J Botany Stud 2021; 6: 139–146.

[3] Shirooie S, Khaledi E, Dehpour AR, Noori T, Khazaei M, Sadeghi F, et al. The effect of dapsone in testosterone enanthate-induced polycystic ovary syndrome in rat. J Steroid Biochem Mol Biol 2021; 214: 105977.

[4] Mahmoud AA, Elfiky AM, Abo-Zeid FS. The anti-androgenic effect of quercetin on hyperandrogenism and ovarian dysfunction induced in a dehydroepiandrosterone rat model of polycystic ovary syndrome. Steroids 2022; 177: 108936.

[5] Salahi E, Amidi F, Zahiri Z, Aghahosseini M, Mashayekhi F, Amani Abkenari S, et al. The effect of mitochondria-targeted antioxidant MitoQ10 on redox signaling pathway components in PCOS mouse model. Arch Gynecol Obstet 2022; 305: 985–994.

[6] Zhang Y, Hu M, Meng F, Sun X, Xu H, Zhang J, et al. Metformin ameliorates uterine defects in a rat model of polycystic ovary syndrome. EBioMedicine 2017; 18: 157–170.

[7] Sasaki H, Hamatani T, Kamijo Sh, Iwai M, Kobanawa M, Ogawa S, et al. Impact of oxidative stress on age-associated decline in oocyte developmental competence. Front Endocrinol 2019; 10: 811.

[8] Cruz-Sanabria F, Carmassi C, Bruno S, Bazzani A, Carli M, Scarselli M, et al. Melatonin as a chronobiotic with sleep-promoting properties. Curr Neuropharmacol 2022; in Press.

[9] Mojaverrostami S, Asghari N, Khamisabadi M, Heidari Khoei H. The role of melatonin in polycystic ovary syndrome: A review. Int J Reprod BioMed 2019; 17: 865–882.

[10] Ahmadi M, Rostamzadeh A, Fathi F, Mohammadi M, Rezaie MJ. The effect of Melatonin on histological changes of ovary in induced polycystic ovary syndrome model in mice. Middle East Fertil Soc J 2017; 22: 255– 259.

[11] Zheng J-H, Zhang J-K, Tian Y-P, Song Y-B, Yang ZW, Huang X-H. A stereological study of mouse ovary tissues for 3D bioprinting application. Cell Mol Bioeng 2021; 14: 259–265.

[12] Sherafatmanesh S, Ekramzadeh M, Tanideh N, Golmakani M-T, Koohpeyma F. The effects of thylakoidrich spinach extract and aqueous extract of caraway (Carum carvi L.) in letrozole-induced polycystic ovarian syndrome rats. BMC Complement Med Ther 2020; 20: 249.

[13] Noorafshan A, Ahmadi M, Mesbah S-F, Karbalay-Doust S. Stereological study of the effects of letrozole and estradiol valerate treatment on the ovary of rats. Clin Exp Reprod Med 2013; 40: 115–121.

[14] Namavar MR, Ghalavandi M, Bahmanpour S. The effect of glutathione and buserelin on the stereological parameters of the hypothalamus in the cyclophosphamide-treated mice. J Chem Neuroanat 2020; 110: 101871.

[15] Ashkani-Esfahani S, Koohi Hosseinabadi O, Moezzi P, Moafpourian Y, Kardeh S, Rafiee S, et al. Verapamil, a calcium-channel blocker, improves the wound healing process in rats with excisional full-thickness skin wounds based on stereological parameters. Adv Skin Wound Care 2016; 29: 271–274.

[16] Sá SI, Maia J, Bhowmick N, Silva SM, Silva A, Correiada- Silva G, et al. Uterine histopathological changes induced by acute administration of tamoxifen and its modulation by sex steroid hormones. Toxicol Appl Pharmacol 2019; 363: 88–97.

[17] Tanideh R, Delavari Sh, Farshad O, Irajie C, Javad Yavari Barhaghtalab M, Koohpeyma F, et al. Effect of flaxseed oil on biochemical parameters, hormonal indexes and stereological changes in ovariectomized rats. Vet Med Sci 2021; 7: 521–533.

[18] Kleinert M, Clemmensen C, Hofmann SM, Moore MC, Renner S, Woods SC, et al. Animal models of obesity and diabetes mellitus. Nat Rev Endocrinol 2018; 14: 140–162.

[19] Kelley ST, Skarra DV, Rivera AJ, Thackray VG. The gut microbiome is altered in a letrozole-induced mouse model of polycystic ovary syndrome. PloS One 2016; 11: e0146509.

[20] Kauffman AS, Thackray VG, Ryan GE, Tolson KP, Glidewell-Kenney CA, Semaan SJ, et al. A novel letrozole model recapitulates both the reproductive and metabolic phenotypes of polycystic ovary syndrome in female mice. Biol Reprod 2015; 93: 69.

[21] Skarra DV, Hernandez-Carretero A, Rivera AJ, Anvar AR, Thackray VG. Hyperandrogenemia induced by letrozole treatment of pubertal female mice results in hyperinsulinemia prior to weight gain and insulin resistance. Endocrinology 2017; 158: 2988–3003.

[22] Bertoldo MJ, Caldwell AS, Riepsamen AH, Lin D, Gonzalez MB, Robker RL, et al. A hyperandrogenic environment causes intrinsic defects that are detrimental to follicular dynamics in a PCOS mouse model. Endocrinology 2019; 160: 699–715.

[23] Zhou L, Han X, Li W, Wang N, Yao L, Zhao Y, et al. N6-methyladenosine demethylase FTO induces the dysfunctions of ovarian granulosa cells by upregulating flotillin 2. Reprod Sci 2022; 29: 1305–1315.

[24] 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: 21.

[25] Seymen CM, Yar Saglam AS, Elmazoglu Z, Arik GN Take Kaplanoglu G. Involvement of endometrial IGF- 1R/IGF-1/Bcl-2 pathways in experimental polycystic ovary syndrome: Identification of the regulatory effect of melatonin. Tissue Cell 2021; 73: 101585.

[26] Ferreira SR, Goyeneche AA, Heber MF, Abruzzese GA, Telleria CM, Motta AB. Prenatally androgenized female rats develop uterine hyperplasia when adult. Mol Cell Endocrinol 2020; 499: 110610.

[27] Bracho GS, Acosta MV, Altamirano GA, Tschopp MV, Luque EH, Kass l, et al. Androgen receptor and uterine histoarchitecture in a PCOS rat model. Mol Cell Endocrinol 2020; 518: 110973.

[28] Bao L, Syed R, Aloahd MS. Analysis of VEGF gene polymorphisms and serum VEGF protein levels contribution in polycystic ovary syndrome of patients. Mol Biol Rep 2019; 46: 5821–5829.

[29] Guan Y, Wang D, Bu H, Zhao T, Wang H. The effect of metformin on polycystic ovary syndrome in overweight women: A systematic review and meta-analysis of randomized controlled trials. Int J Endocrinol 2020; 2020: 5150684.

[30] Mahamed RR, Maganhin CC, Sasso GRS, de Jesus Simoes M, Baracat MCP, Baracat EC, et al. Metformin improves ovarian follicle dynamics by reducing theca cell proliferation and CYP-17 expression in an androgenized rat model. J Ovarian Res 2018; 11: 18.

[31] Rencber SF, Ozbek SK, Eraldemir C, Sezer Z, Kum T, Ceylan S, et al. Effect of resveratrol and metformin on ovarian reserve and ultrastructure in PCOS: An experimental study. J Ovarian Res 2018; 11: 55.

[32] Guo M, Zhou JJ, Huang W. Metformin alleviates endometrial hyperplasia through the UCA1/miR‑144/TGF‑β1/AKT signaling pathway. Int J Mol Med 2020; 45: 623–633.

[33] Yu K, Wang R-X, Li M-H, Sun T-C, Zhou Y-W, Li YY, et al. Melatonin reduces androgen production and upregulates heme oxygenase-1 expression in granulosa cells from PCOS patients with hypoestrogenia and hyperandrogenia. Oxid Med Cell Longev 2019; 2019: 8218650.

[34] Mousavi R, Alizadeh M, Asghari Jafarabadi M, Heidari L, Nikbakht R, Babaahmadi Rezaei H, et al. Effects of melatonin and/or magnesium supplementation on biomarkers of inflammation and oxidative stress in women with polycystic ovary syndrome: A randomized, double-blind, placebo-controlled trial. Biol Trace Elem Res 2022; 200: 1010–1019.

[35] Lombardi LA, Mattos LSd, Simoes RS, Florencio-Silva R, Sasso GRdS, Carbonel AAF, et al. Melatonin may prevent or reverse polycystic ovary syndrome in rats. Rev Assoc Med Bras 2019; 65: 1008–1014.

[36] Hansda SR, Haldar Ch. Uterine anomalies in cell proliferation, energy homeostasis and oxidative stress in PCOS hamsters, M. auratus: Therapeutic potentials of melatonin. Life Sci 2021; 281: 119755.

[37] de Almeida Chuffa LG, Lupi LA, Cucielo MS, Silveira HS, Reiter RJ, Seiva FR. Melatonin promotes uterine and placental health: Potential molecular mechanisms. Int J Mol Sci 2020; 21: 300.

[38] Ma Q, Reiter RJ, Chen Y. Role of melatonin in controlling angiogenesis under physiological and pathological conditions. Angiogenesis 2020; 23: 91–104.