The role of melatonin in polycystic ovary syndrome: A review

Sina Mojaverrostami, Narjes Asghari, Mahsa Khamisabadi, Heidar Heidari Khoei, Sina Mojaverrostami, Narjes Asghari, Mahsa Khamisabadi, Heidar Heidari Khoei

Abstract

Background: Polycystic ovary syndrome (PCOS) is a widespread endocrine disorder, affecting approximately 20% of women within reproductive age. It is associated with hyperandrogenism, obesity, menstrual irregularity, and anovulatory infertility. Melatonin is the main pineal gland hormone involved in the regulation of the circadian rhythm. In recent years, it has been observed that a reduction in melatonin levels of follicular fluid exists in PCOS patients. Melatonin receptors in the ovary and intra-follicular fluid adjust sex steroid secretion at different phases of ovarian follicular maturation. Moreover, melatonin is a strong antioxidant and an effective free radical scavenger, which protects ovarian follicles during follicular maturation.

Objective: In this paper, we conducted a literature review and the summary of the current research on the role of melatonin in PCOS.

Materials and methods: Electronic databases including PubMed/MEDLINE, Web of Science, Scopus, and Reaxys were searched from their inception to October 2018 using the keywords "Melatonin" AND "Polycystic ovary syndrome" OR "PCOS."

Results: Based on the data included in our review, it was found that the administration of melatonin can improve the oocyte and embryo quality in PCOS patients. It may also have beneficial effects in correcting the hormonal alterations in PCOS patients.

Conclusion: Since metabolic dysfunction is the major finding contributing to the initiation of PCOS, melatonin can hinder this process via its improving effects on metabolic functions.

Keywords: Infertility; Melatonin; PCOS.; Hyperandrogenism.

Conflict of interest statement

All authors declare no conflicts of interest.

Copyright © 2019 Mojaverrostami et al.

Figures

Figure 1
Figure 1
Flow diagram of the search strategy.
Figure 2
Figure 2
The role of melatonin in the pathogenesis of PCOS.

References

    1. Shaikh N., Dadachanji R., Mukherjee S. Genetic markers of polycystic ovary syndrome: emphasis on insulin resistance. International Journal of Medical Genetics. 2014;2014 doi: 10.1155/2014/478972.478972
    1. The Rotterdam ESHRE/ASRM-Sponsored PCOS Consensus Workshop Group Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome (PCOS) Human Reproduction. 2004;19(1):41–47. doi: 10.1093/humrep/deh098.
    1. Azziz R., Carmina E., Dewailly D., Diamanti-Kandarakis E., Escobar-Morreale H. F., Futterweit W., Janssen O. E., Legro R. S., Norman R. J., Taylor A. E., Witchel S. F. The Androgen Excess and PCOS Society criteria for the polycystic ovary syndrome: the complete task force report. Fertility and Sterility. 2009;91(2):456–488. doi: 10.1016/j.fertnstert.2008.06.035.
    1. DEUGARTE C., BARTOLUCCI A., AZZIZ R. Prevalence of insulin resistance in the polycystic ovary syndrome using the homeostasis model assessment. Fertility and Sterility. 2005;83(5):1454–1460. doi: 10.1016/j.fertnstert.2004.11.070.
    1. Dunaif A. Perspectives in Polycystic Ovary Syndrome: From Hair to Eternity. The Journal of Clinical Endocrinology & Metabolism. 2016;101(3):759–768. doi: 10.1210/jc.2015-3780.
    1. Heidari Khoei H., Dehdehi L., Moloudizargari M., Baninameh Z., Rezaie-Chamani S., Gea Conti O. Female obesity and clinical outcomes of assisted reproductive technologies (ART): An updated systematic review and Meta-Analysis. Int J Med Res Health Sci. 2016;5:157–170.
    1. Morgante G., Massaro M. G., Di Sabatino A., Cappelli V., De Leo V. Therapeutic approach for metabolic disorders and infertility in women with PCOS. Gynecological Endocrinology. 2017;34(1):4–9. doi: 10.1080/09513590.2017.1370644.
    1. Sirmans S. M., Pate K. A. Epidemiology, diagnosis, and management of polycystic ovary syndrome. Journal of Clinical Epidemiology. 2013;6(1):1–13. doi: 10.2147/clep.s37559.
    1. Asghari M. H., Moloudizargari M., Bahadar H., Abdollahi M. A review of the protective effect of melatonin in pesticide-induced toxicity. Expert Opinion on Drug Metabolism & Toxicology. 2017;13(5):545–554. doi: 10.1080/17425255.2016.1214712.
    1. Asghari M. H., Moloudizargari M., Ghobadi E., Fallah M., Abdollahi M. Melatonin as a multifunctional anti-cancer molecule: Implications in gastric cancer. Life Sciences. 2017;185:38–45. doi: 10.1016/j.lfs.2017.07.020.
    1. Asghari M. H., Moloudizargari M., Baeeri M., Baghaei A., Rahimifard M., Solgi R., Jafari A., Aminjan H. H., Hassani S., Moghadamnia A. A., Ostad S. N., Abdollahi M. On the mechanisms of melatonin in protection of aluminum phosphide cardiotoxicity. Archives of Toxicology. 2017;91(9):3109–3120. doi: 10.1007/s00204-017-1998-6.
    1. Reiter R. J., Tan D., Tamura H., Cruz M. H., Fuentes-Broto L. Clinical relevance of melatonin in ovarian and placental physiology: a review. Gynecological Endocrinology. 2013;30(2):83–89. doi: 10.3109/09513590.2013.849238.
    1. Asghari M. H., Abdollahi M., de Oliveira M. R., Nabavi S. M. A review of the protective role of melatonin during phosphine-induced cardiotoxicity: focus on mitochondrial dysfunction, oxidative stress and apoptosis. Journal of Pharmacy and Pharmacology. 2017;69(3):236–243. doi: 10.1111/jphp.12682.
    1. Moradkhani F., Moloudizargari M., Fallah M., Asghari N., Heidari Khoei H., Asghari M. H. Immunoregulatory role of melatonin in cancer. Journal of Cellular Physiology. 2019;235(2):745–757. doi: 10.1002/jcp.29036.
    1. Moloudizargari M., Moradkhani F., Asghari N., Fallah M., Asghari M. H., Moghadamnia A. A., Abdollahi M. NLRP inflammasome as a key role player in the pathogenesis of environmental toxicants. Life Sciences. 2019;231:116585. doi: 10.1016/j.lfs.2019.116585.
    1. Goradel N. H., Asghari M. H., Moloudizargari M., Negahdari B., Haghi-Aminjan H., Abdollahi M. Melatonin as an angiogenesis inhibitor to combat cancer: Mechanistic evidence. Toxicology and Applied Pharmacology. 2017;335:56–63. doi: 10.1016/j.taap.2017.09.022.
    1. Berson D. M., Dunn F. A., Takao M. Phototransduction by retinal ganglion cells that set the circadian clock. Science. 2002;295(5557):1070–1073. doi: 10.1126/science.1067262.
    1. Kohsaka A., Bass J. A sense of time: how molecular clocks organize metabolism. Trends in Endocrinology & Metabolism. 2007;18(1):4–11. doi: 10.1016/j.tem.2006.11.005.
    1. Reiter R. J., Tan D.-X., Manchester L. C., Paredes S. D., Mayo J. C., Sainz R. M. Melatonin and reproduction revisited. Biology of Reproduction. 2009;81(3):445–456. doi: 10.1095/biolreprod.108.075655.
    1. Tan D.-X., Manchester L. C., Hardeland R., Lopez-Burillo S., Mayo J. C., Sainz R. M., Reiter R. J. Melatonin: a hormone, a tissue factor, an autocoid, a paracoid, and an antioxidant vitamin. Journal of Pineal Research. 2003;34(1):75–78. doi: 10.1034/j.1600-079x.2003.02111.x.
    1. Itoh M. T., Ishizuka B., Kudo Y., Fusama S., Amemiya A., Sumi Y. Detection of melatonin and serotonin N-acetyltransferase and hydroxyindole-O-methyltransferase activities in rat ovary. Molecular and Cellular Endocrinology. 1997;136(1):7–13. doi: 10.1016/S0303-7207(97)00206-2.
    1. Reiter R. J., Tamura H., Tan D. X., Xu X. Melatonin and the circadian system: contributions to successful female reproduction. Fertility and Sterility. 2014;102(2):321–328. doi: 10.1016/j.fertnstert.2014.06.014.
    1. Majumdar A., Sharma MN. in. Kamini AR. Principles Practice of Assisted Reproductive Technology. New Delhi, India: Jaypee Brothers Medical Publishers LTD; 2014. Anovulatory Infertility.
    1. McCartney C. R., Eagleson C. A., Marshall J. C. Regulation of gonadotropin secretion: implications for polycystic ovary syndrome. Seminars in Reproductive Medicine. 2002;20(4):317–325. doi: 10.1055/s-2002-36706.
    1. Johansson Julia, Stener-Victorin Elisabet. Polycystic Ovary Syndrome: Effect and Mechanisms of Acupuncture for Ovulation Induction. Evidence-Based Complementary and Alternative Medicine. 2013;2013 doi: 10.1155/2013/762615.762615
    1. Kelly C. J., Stenton S. R., Lashen H. Insulin-like growth factor binding protein-1 in PCOS: a systematic review and meta-analysis. Human Reproduction Update. 2010;17(1):4–16. doi: 10.1093/humupd/dmq027.
    1. Azziz R., Black V. Y., Knochenhauer E. S., Hines G. A., Boots L. R. Ovulation after glucocorticoid suppression of adrenal androgens in the polycystic ovary syndrome is not predicted by the basal dehydroepiandrosterone sulfate level. The Journal of Clinical Endocrinology & Metabolism. 1999;84(3):946–950. doi: 10.1210/jc.84.3.946.
    1. Fassnacht M., Schlenz N., Schneider S. B., Wudy S. A., Allolio B., Arlt W. Beyond Adrenal and Ovarian Androgen Generation: Increased Peripheral 5α-Reductase Activity in Women with Polycystic Ovary Syndrome. The Journal of Clinical Endocrinology & Metabolism. 2003;88(6):2760–2766. doi: 10.1210/jc.2002-021875.
    1. Polak K., Czyzyk A., Simoncini T., Meczekalski B. New markers of insulin resistance in polycystic ovary syndrome. Journal of Endocrinological Investigation. 2017;40(1) doi: 10.1007/s40618-016-0523-8.
    1. Corbould A. Chronic testosterone treatment induces selective insulin resistance in subcutaneous adipocytes of women. Journal of Endocrinology. 2007;192(3):585–594. doi: 10.1677/joe.1.07070.
    1. Diamanti-Kandarakis E. Role of obesity and adiposity in polycystic ovary syndrome. International Journal of Obesity. 2007;31(supplement 2):S8–S13. doi: 10.1038/sj.ijo.0803730.
    1. Tamura H., Nakamura Y., Korkmaz A., Manchester L. C., Tan D.-X., Sugino N., Reiter R. J. Melatonin and the ovary: physiological and pathophysiological implications. Fertility and Sterility. 2009;92(1):328–343. doi: 10.1016/j.fertnstert.2008.05.016.
    1. Luboshitzky R., Shen-Orr Z., Herer P., Nave R. Urinary 6-sulfatoxymelatonin excretion in hyperandrogenic women with polycystic ovary syndrome: the effect of ethinyl estradiol–cyproterone acetate treatment. Gynecological Endocrinology. 2009;17(6):441–447. doi: 10.1080/09513590312331290368.
    1. Luboshitzky R., Herer P., Shen-Orr Z. Urinary 6-Sulfatoxymelatonin Excretion in Hyperandrogenic Women: The Effect of Cyproterone Acetate-Ethinyl Estradiol Treatment. Experimental and Clinical Endocrinology & Diabetes. 2004;112(02):102–107. doi: 10.1055/s-2004-815765.
    1. Luboshitzky R., Qupti G., Ishay A., Shen-Orr Z., Futerman B., Linn S. Increased 6-sulfatoxymelatonin excretion in women with polycystic ovary syndrome. Fertility and Sterility. 2001;76(3):506–510. doi: 10.1016/S0015-0282(01)01930-6.
    1. Daya S., Potgieter B. The effect of castration, testosterone and estradiol on14C-serotonin metabolism by organ cultures of male rat pineal glands. Experientia. 1985;41(2):275–276. doi: 10.1007/BF02002634.
    1. Hayashi K., Okatani Y. Mechanisms underlying the effects of estrogen on nocturnal melatonin synthesis in peripubertal female rats: Relation to norepinephrine and adenylate cyclase. Journal of Pineal Research. 1999;26(3):178–183. doi: 10.1111/j.1600-079X.1999.tb00581.x.
    1. Kruijver F. P., Swaab D. F. Sex Hormone Receptors Are Present in the Human Suprachiasmatic Nucleus. Neuroendocrinology. 2002;75(5):296–305. doi: 10.1159/000057339.
    1. Tanavde V. S., Maitra A. In Vitro Modulation of Steroidogenesis and Gene Expression by Melatonin: A Study with Porcine Antral Follicles. Endocrine Research. 2009;29(4):399–410. doi: 10.1081/ERC-120026946.
    1. Jain M., Jain S., Singh T., Haldar C., Jain P. Melatonin and its correlation with testosterone in polycystic ovarian syndrome. Journal of Human Reproductive Sciences. 2013;6(4):253. doi: 10.4103/0974-1208.126295.
    1. Andreeva E. N., Absatarova Y. S., Sheremetyeva E. V., Derkach D. A., Ponomareva T. A., Tiulpakov A. N., Ioutsi V. A., Ilyin A. V., Murvatov K. D. Analysis of the informativeness of melatonin evaluation in polycystic ovary syndrome. Obesity and Metabolism. 2016;13(4):15–20.
    1. Shreeve N., Cagampang F., Sadek K., Tolhurst M., Houldey A., Hill C. M., Brook N., MacKlon N., Cheong Y. Poor sleep in PCOS; is melatonin the culprit? Human Reproduction. 2013;28(5):1348–1353. doi: 10.1093/humrep/det013.
    1. Basheer M., Rai S., Hsu T. Melatonin vs. phytomelatonin: Therapeutic uses with special reference to polycystic ovarian syndrome (PCOS) Cogent Biology. 2016;2(1) doi: 10.1080/23312025.2015.1136257.
    1. González F., Rote N. S., Minium J., Kirwan J. P. Reactive oxygen species-induced oxidative stress in the development of insulin resistance and hyperandrogenism in polycystic ovary syndrome. The Journal of Clinical Endocrinology & Metabolism. 2006;91(1):336–340. doi: 10.1210/jc.2005-1696.
    1. Sabuncu T., Vural H., Harma M., Harma M. Oxidative stress in polycystic ovary syndrome and its contribution to the risk of cardiovascular disease. Clinical Biochemistry. 2001;34(5):407–413. doi: 10.1016/s0009-9120(01)00245-4.
    1. Gupta R. K., Miller K. P., Babus J. K., Flaws J. A. Methoxychlor inhibits growth and induces atresia of antral follicles through an oxidative stress pathway. Toxicological Sciences. 2006;93(2):382–389. doi: 10.1093/toxsci/kfl052.
    1. Reiter R. J., Tan D., Maldonado M. D. Melatonin as an antioxidant: physiology versus pharmacology. Journal of Pineal Research. 2005;39(2):215–216. doi: 10.1111/j.1600-079X.2005.00261.x.
    1. Tan D., Reiter R. J., Manchester L. C., Yan M., El-Sawi M., Sainz R. M., Mayo J. C., Kohen R., Allegra M., Hardeland R. Chemical and physical properties and potential mechanisms: melatonin as a broad spectrum antioxidant and free radical scavenger. Current Topics in Medicinal Chemistry. 2002;2(2):181–197. doi: 10.2174/1568026023394443.
    1. Terzieva D. D., Orbetzova M. M., Mitkov M. D., Mateva N. G. Serum melatonin in women with polycystic ovary syndrome. Folia Medica. 2013;55(2):10–15. doi: 10.2478/folmed-2013-0012.
    1. Zafari Zangeneh F., Naghizadeh MM., Abdollahi A., Bagheri M. Synchrony between ovarian function sleep in polycystic ovary syndrome patients. Open Journal of Obstetrics and Gynecology. 2014;4:725–731. doi: 10.4236/ojog.2014.412101.
    1. Wang L., Wang Y., Zhang X., Shi J., Wang M., Wei Z., Zhao A., Li B., Zhao X., Xing Q. Common genetic variation in MTNR1B is associated with serum testosterone, glucose tolerance, and insulin secretion in polycystic ovary syndrome patients. Fertility and Sterility. 2010;94(6):2486–2489.e2. doi: 10.1016/j.fertnstert.2010.01.059.
    1. Li C., Shi Y., You L., Wang L., Chen Z. Melatonin Receptor 1A Gene Polymorphism Associated with Polycystic Ovary Syndrome. Gynecologic and Obstetric Investigation. 2011;72(2):130–134. doi: 10.1159/000323542.
    1. Li C., Shi Y., You L., Wang L., Chen Z. Association of rs10830963 and rs10830962 SNPs in the melatonin receptor (MTNR1B) gene among Han Chinese women with polycystic ovary syndrome. Molecular Human Reproduction. 2011;17(3):193–198. doi: 10.1093/molehr/gaq087.
    1. Song X., Sun X., Ma G., Sun Y., Shi Y., Du Y., Chen Z. Family association study between melatonin receptor gene polymorphisms and polycystic ovary syndrome in Han Chinese. European Journal of Obstetrics & Gynecology and Reproductive Biology. 2015;195:108–112. doi: 10.1016/j.ejogrb.2015.09.043.
    1. Peschke E., Frese T., Chankiewitz E., Peschke D., Preiss U., Schneyer U., Spessert R., Mühlbauer E. Diabetic Goto Kakizaki rats as well as type 2 diabetic patients show a decreased diurnal serum melatonin level and an increased pancreatic melatonin-receptor status. Journal of Pineal Research. 2006;40(2):135–143. doi: 10.1111/j.1600-079X.2005.00287.x.
    1. Pai S. A., Majumdar A. S. Protective effects of melatonin against metabolic and reproductive disturbances in polycystic ovary syndrome in rats. Journal of Pharmacy and Pharmacology. 2014;66(12):1710–1721. doi: 10.1111/jphp.12297.
    1. Faria J. A., Kinote A., Ignacio-Souza L. M., de Araújo T. M., Razolli D. S., Doneda D. L., Paschoal L. B., Lellis-Santos C., Bertolini G. L., Velloso L. A., Bordin S., Anhê G. F. Melatonin acts through MT1/MT2 receptors to activate hypothalamic Akt and suppress hepatic gluconeogenesis in rats. American Journal of Physiology-Endocrinology and Metabolism. 2013;305(2):E230–E242. doi: 10.1152/ajpendo.00094.2013.
    1. Ha E., Yim S.-V., Chung J.-H., Yoon K.-S., Kang I., Yong H. C., Baik H. H. Melatonin stimulates glucose transport via insulin receptor substrate-1/phosphatidylinositol 3-kinase pathway in C2C12 murine skeletal muscle cells. Journal of Pineal Research. 2006;41(1):67–72. doi: 10.1111/j.1600-079X.2006.00334.x.
    1. Chottanapund S., Van Duursen M., Navasumrit P., Hunsonti P., Timtavorn S., Ruchirawat M., Van den Berg M. Anti-aromatase effect of resveratrol and melatonin on hormonal positive breast cancer cells co-cultured with breast adipose fibroblasts. Toxicology in Vitro. 2014;28(7):1215–1221. doi: 10.1016/j.tiv.2014.05.015.
    1. Al-Qadhi H. I. Effect of melatonin supplementation on serum lh level and bmi in women with polycystic ovarian syndrome. Journal of Pharmaceutical Sciences and Research. 2018;10(1):1–4.
    1. Swanton A., Storey L., McVeigh E., Child T. IVF outcome in women with PCOS, PCO and normal ovarian morphology. European Journal of Obstetrics & Gynecology and Reproductive Biology. 2010;149(1):68–71. doi: 10.1016/j.ejogrb.2009.11.017.
    1. Rama Raju G., Chavan R., Deenadayal M., Govindarajan M., Gunasheela D., Gutgutia R., Haripriya G., Patel N., Patki A. Luteinizing hormone and follicle stimulating hormone synergy: A review of role in controlled ovarian hyper-stimulation. Journal of Human Reproductive Sciences. 2013;6(4):227. doi: 10.4103/0974-1208.126285.
    1. Doronzo G., Russo I., Mattiello L., Anfossi G., Bosia A., Trovati M. Insulin activates vascular endothelial growth factor in vascular smooth muscle cells: influence of nitric oxide and of insulin resistance. European Journal of Clinical Investigation. 2004;34(10):664–673. doi: 10.1111/j.1365-2362.2004.01412.x.
    1. Pacchiarotti A., Carlomagno G., Antonini G., Pacchiarotti A. Effect of myo-inositol and melatonin versus myo-inositol, in a randomized controlled trial, for improving in vitro fertilization of patients with polycystic ovarian syndrome. Gynecological Endocrinology. 2016;32(1):69–73. doi: 10.3109/09513590.2015.1101444.
    1. Rose B. I. Avoidance of severe ovarian hyperstimulation with IVM treatment. Development of In Vitro Maturation for Human Oocytes: Natural and Mild Approaches to Clinical Infertility Treatment. 2017:317–327.
    1. Virant-Klun Irma, Krijgsveld Jeroen, Huntriss John, Rodgers Raymond J. New Advances in Reproductive Biomedicine. BioMed Research International. 2014;2014 doi: 10.1155/2014/529170.529170
    1. Hao Y., Zhang Z., Han D., Cao Y., Zhou P., Wei Z., Lv M., Chen D. Gene expression profiling of human blastocysts from in vivo and ‘rescue IVM’ with or without melatonin treatment. Molecular Medicine Reports. 2017;16(2):1278–1288. doi: 10.3892/mmr.2017.6742.
    1. Lemos-Jordão A. J., Costa F. S., Peixoto C. A., Teixeira Á. A., Silva S. B., Ferreira C. G., Oliveira A. G., Luna R. L., Silva E. P., D’assunção C. G., Wanderley-Teixeira V. Combination of Melatonin and Metformin Hydrochloride for Treatment Polycystic Ovarian in Female Rats. Acta Scientiae Veterinariae. 2018;44(1):10. doi: 10.22456/1679-9216.81208.
    1. Fernando S., Wallace E. M., Vollenhoven B., Lolatgis N., Hope N., Wong M., Lawrence M., Lawrence A., Russell C., Leong K., Thomas P., Rombauts L. Melatonin in Assisted Reproductive Technology: A Pilot Double-Blind Randomized Placebo-Controlled Clinical Trial. Frontiers in Endocrinology. 2018;9 doi: 10.3389/fendo.2018.00545.
    1. Tamura H., Takasaki A., Taketani T., Tanabe M., Kizuka F., Lee L., Tamura I., Maekawa R., Asada H., Yamagata Y., Sugino N. Melatonin as a free radical scavenger in the ovarian follicle. Endocrine Journal. 2013;60(1):1–13. doi: 10.1507/endocrj.EJ12-0263.
    1. Najafabadi M. E., Malekshah A. K., Moghaddam A. E., Hamidabadi H. G., Heidari M. Effect of embryonic fibroblast cells conditioned medium on in vitro maturation of immature mouse oocytes. Journal of Mazandaran University of Medical Sciences. 2014;24(1):106–114.
    1. Kim M. K., Park E. A., Kim H. J., Choi W. Y., Cho J. H., Lee W. S., Cha K. Y., You Kim S., Lee D. R., Yoon T. K. Does supplementation of in-vitro culture medium with melatonin improve IVF outcome in PCOS? Reproductive BioMedicine Online. 2013;26(1):22–29. doi: 10.1016/j.rbmo.2012.10.007.
    1. Nikmard F., Hosseini E., Bakhtiyari M., Ashrafi M., Amidi F., Aflatoonian R. Effects of melatonin on oocyte maturation in PCOS mouse model. Animal Science Journal. 2017;88(4):586–592. doi: 10.1111/asj.12675.
    1. Ahmadi M., Rostamzadeh A., Fathi F., Mohammadi M., Rezaie M. J. The effect of Melatonin on histological changes of ovary in induced polycystic ovary syndrome model in mice. Middle East Fertility Society Journal. 2017;22(4):255–259. doi: 10.1016/j.mefs.2017.03.009.
    1. Rocha R., Lima L., Alves A., Celestino J., Matos M., Lima-Verde I., Bernuci M., Lopes C., Báo S., Campello C., Rodrigues A., Figueiredo J. Interaction between melatonin and follicle-stimulating hormone promotes in vitro development of caprine preantral follicles. Domestic Animal Endocrinology. 2013;44(1):1–9. doi: 10.1016/j.domaniend.2012.07.001.
    1. Galano A., Tan D. X., Reiter R. J. Melatonin as a natural ally against oxidative stress: a physicochemical examination. Journal of Pineal Research. 2011;51(1):1–16. doi: 10.1111/j.1600-079X.2011.00916.x.
    1. Polson D., Wadsworth J., Adams J., Franks S. POLYCYSTIC OVARIES—A COMMON FINDING IN NORMAL WOMEN. The Lancet. 1988;331(8590):870–872. doi: 10.1016/S0140-6736(88)91612-1.
    1. Kang Xuezhi, Jia Lina, Shen Xueyong. Manifestation of Hyperandrogenism in the Continuous Light Exposure-Induced PCOS Rat Model. BioMed Research International. 2015;2015 doi: 10.1155/2015/943694.943694
    1. Prata Lima M., Baracat E., Simões M. Effects of melatonin on the ovarian response to pinealectomy or continuous light in female rats: similarity with polycystic ovary syndrome. Brazilian Journal of Medical and Biological Research. 2004;37(7):987–995. doi: 10.1590/S0100-879X2004000700007.
    1. Basheer M., Rai S., Ghosh H., Ahmad Haja Y. Therapeutic Efficacy of Melatonin Against Polycystic Ovary Syndrome (PCOS) Induced by Letrozole in Wistar Rat. Pakistan Journal of Biological Sciences. 2018;21(7):340–347. doi: 10.3923/pjbs.2018.340.347.
    1. Giannetto A., Fernandes J. M., Nagasawa K., Mauceri A., Maisano M., De Domenico E., Cappello T., Oliva S., Fasulo S. Influence of continuous light treatment on expression of stress biomarkers in Atlantic cod. Developmental & Comparative Immunology. 2014;44(1):30–34. doi: 10.1016/j.dci.2013.11.011.
    1. Fenkci V., Fenkci S., Yilmazer M., Serteser M. Decreased total antioxidant status and increased oxidative stress in women with polycystic ovary syndrome may contribute to the risk of cardiovascular disease. Fertility and Sterility. 2003;80(1):123–127. doi: 10.1016/s0015-0282(03)00571-5.
    1. Lemos A. J., Peixoto C. A., Teixeira Á. A., Luna R. L., Rocha S. W., Santos H. M., Silva A. K., Nunes A. K., Wanderley-Teixeira V. Effect of the combination of metformin hydrochloride and melatonin on oxidative stress before and during pregnancy, and biochemical and histopathological analysis of the livers of rats after treatment for polycystic ovary syndrome. Toxicology and Applied Pharmacology. 2014;280(1):159–168. doi: 10.1016/j.taap.2014.05.015.
    1. Kim J.-B., Jung J. Y., Ahn J.-C., Rhee C. K., Hwang H. Antioxidant and anti-apoptotic effect of melatonin on the vestibular hair cells of rat utricles. Clinical and Experimental Otorhinolaryngology. 2009;2(1):6–12. doi: 10.3342/ceo.2009.2.1.6.
    1. Kang J.-T., Koo O.-J., Kwon D.-K., Park H.-J., Jang G., Kang S.-K., Lee B.-C. Effects of melatonin on in vitro maturation of porcine oocyte and expression of melatonin receptor RNA in cumulus and granulosa cells. Journal of Pineal Research. 2009;46(1):22–28. doi: 10.1111/j.1600-079X.2008.00602.x.
    1. Korkmaz A., Rosales-Corral S., Reiter R. J. Gene regulation by melatonin linked to epigenetic phenomena. Gene. 2012;503(1):1–11. doi: 10.1016/j.gene.2012.04.040.
    1. Shkolnik K., Tadmor A., Ben-Dor S., Nevo N., Galiani D., Dekel N. Reactive oxygen species are indispensable in ovulation. Proceedings of the National Acadamy of Sciences of the United States of America. 2011;108(4):1462–1467. doi: 10.1073/pnas.1017213108.
    1. Shi J., Tian X., Zhou G., Wang L., Gao C., Zhu S., Zeng S., Tian J., Liu G. Melatonin exists in porcine follicular fluid and improves in vitro maturation and parthenogenetic development of porcine oocytes. Journal of Pineal Research. 2009;47(4):318–323. doi: 10.1111/j.1600-079X.2009.00717.x.
    1. van Hooff M. H., Voorhorst F. J., Kaptein M. B., Hirasing R. A., Koppenaal C., Schoemaker J. Polycystic ovaries in adolescents and the relationship with menstrual cycle patterns, luteinizing hormone, androgens, and insulin. Fertility and Sterility. 2000;74(1):49–58. doi: 10.1016/S0015-0282(00)00584-7.
    1. Dowdy D. Emotional Needs of Teens With Polycystic Ovary Syndrome. Journal of Pediatric Nursing. 2012;27(1):55–64. doi: 10.1016/j.pedn.2010.08.001.
    1. Tagliaferri V., Romualdi D., Scarinci E., Cicco S. D., Florio C. D., Immediata V., Tropea A., Santarsiero C. M., Lanzone A., Apa R. Melatonin Treatment May Be Able to Restore Menstrual Cyclicity in Women With PCOS: A Pilot Study. Reproductive Sciences. 2017;25(2):269–275. doi: 10.1177/1933719117711262.
    1. Wilson S. J., Nutt D. J., Alford C., Argyropoulos S. V., Baldwin D. S., Bateson A. N., Britton T. C., Crowe C., Dijk D.-J., Espie C. A., Gringras P., Hajak G., Idzikowski C., Krystal A. D., Nash J. R., Selsick H., Sharpley A. L., Wade A. G. British Association for Psychopharmacology consensus statement on evidence-based treatment of insomnia, parasomnias and circadian rhythm disorders. Journal of Psychopharmacology. 2010;24(11):1577–1601. doi: 10.1177/0269881110379307.
    1. Chan W. Y., Ng T. B. Effects of pineal indoles on ovarian response to gonadotropin-induced ovulation in mice. Journal of Neural Transmission. 1995;100(3):239–246. doi: 10.1007/BF01276461.

Source: PubMed

3
Iratkozz fel