نقش التهاب و استرس اکسیداتیو در سبب‌شناسی سندرم تخمدان پلی‌کیستیک: مطالعه مروری

نوع مقاله : مروری

نویسندگان

1 دکتری تخصصی مهندسی بافت، گروه جنین‌شناسی، مرکز تحقیقات پزشکی تولید مثل، پژوهشکده زیست‌شناسی و علوم پزشکی تولید مثل جهاد دانشگاهی، پژوهشگاه رویان، تهران، ایران.

2 کارشناس ارشد مامایی، گروه اندوکرینولوژی و ناباروری زنان، مرکز تحقیقات پزشکی تولید مثل، پژوهشکده زیست‌شناسی و علوم پزشکی تولید مثل جهاد دانشگاهی، پژوهشگاه رویان، تهران، ایران.

3 دکتری تخصصی آناتومی، گروه جنین‌شناسی، مرکز تحقیقات پزشکی تولید مثل، پژوهشکده زیست‌شناسی و علوم پزشکی تولید مثل جهاد دانشگاهی، پژوهشگاه رویان، تهران، ایران.

چکیده

مقدمه: در سال‌های اخیر نقش التهاب و استرس اکسیداتیو در سبب‌شناسی بیماری سندرم تخمدان پلی‌کیستیک بیش از پیش مورد توجه قرار گرفته است. بررسی مطالعات موجود در رابطه با اثرات واسطه‌های التهابی و استرس اکسیداتیو بر عملکرد تخمدان در بیماران مبتلا به PCOS، حاکی از نقش کلیدی این عوامل در پیشرفت و توسعه عوارض بیماری است. مطالعه مروری حاضر با هدف جمع‌بندی حضور و نقش واسطه‌های التهابی مذکور و همچنین تأثیر رویکردهای درمانی اخیر شامل استفاده از مکمل‌های آنتی‌اکسیدانی و عوامل ضدالتهابی در بهبود نتایج چرخه‌های درمان ناباروری در این بیماران انجام شد.
روشکار: در این مطالعه مروری جهت یافتن مقالات مرتبط، پایگاه‌های اطلاعاتی PubMed، Science Direct، Scopus و Google Scholar با استفاده از کلمات کلیدی: سندرم تخمدان پلی‌کیستیک، التهاب، استرس اکسیداتیو، رادیکال‌های آزاد اکسیژن، مکمل‌های آنتی‌اکسیدانی، درمان ناباروری، آندروژن‌ها، سیتوکین‌ها و مقاومت به انسولین با استفاده از عملگرهای "AND" و "OR" از ژانویه 1991 تا دسامبر 2020 مورد جستجو قرار گرفتند.
یافته ­ها: پس از بررسی تمامی مقالات در این زمینه مشخص شد PCOS به‌عنوان یک بیماری ژنتیکی و خودایمنی با درجاتی از التهاب همراه است که نقش مهمی در سبب‌شناسی، پیشرفت و بروز بیماری دارد. اختلال در تخمک‌گذاری بیمار، افزایش غلظت رادیکال­ های آزاد اکسیژن در سلول­ های گرانولوزا و همچنین مایع فولیکولی در بیماران PCOS نابارور تحت درمان با تکنیک­ های کمک باروری گزارش شده است.
نتیجه ­گیری: به‌نظر می‌رسد هماهنگی بین سه عامل اصلی دریافت کربوهیدرات بالا از طریق رژیم غذایی، مقاومت به انسولین و هیپرآندروژنیسم را می‌توان آغازگر فرآیند القای التهاب و استرس اکسیداتیو در بیماران PCOS دانست. مکمل­ های آنتی‌اکسیدانی و عوامل ضدالتهابی می‌تواند در بهبود نتایج سیکل‌های درمان ناباروری در این بیماران مفید باشد. با این‌حال، جمع‌بندی نتایج هنوز به مطالعات بیشتری نیاز دارد.

کلیدواژه‌ها


عنوان مقاله [English]

The Role of Inflammation and Oxidative Stress in the Etiology of Polycystic Ovary Syndrome: A Review Article

نویسندگان [English]

  • Nahid Nasiri 1
  • Arezoo Arabipoor 2
  • Poopak Eftekhari-Yazdi 3
1 PhD in Tissue Engineering, Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
2 M.Sc. of Midwifery, Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
3 PhD in Anatomy, Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
چکیده [English]

Introduction: In recent years, the role of inflammation and oxidative stress in the etiology of polycystic ovary syndrome has received more attention. Evaluation of the existing studies regarding the effects of inflammatory mediators and oxidative stress on ovarian function in patients with polycystic ovary syndrome (PCOS) indicates the key role of these factors in the progress and development of disease complications. The current review study was conducted with aim to summarize the presence and role of the aforementioned inflammatory mediators as well as the effect of recent treatment approaches including the use of antioxidant supplements and anti-inflammatory agents in improving the results of infertility treatment cycles in these patients.
Methods: In the present review article, to find the related articles, databases of PubMed, Science Direct, Scopus, and Google Scholar databases were searched with the keywords of polycystic ovary syndrome, inflammation, oxidative stress, oxygen-free radicals, antioxidant supplementation, infertility treatment, androgens, cytokines, and insulin resistance using the "AND" and "OR" operators from January 1991 to December 2020.
Results: After reviewing all the articles in this field, it was found that PCOS, as a genetic and autoimmune disease, is associated with low degrees of inflammation, which plays an important role in the etiology, progression and occurrence of the disease. Moreover, ovulatory dysfunction, increased concentrations of oxygen free radicals in granulosa cells, as well as the follicular fluid of infertile PCOS patients under ART treatment, have been reported.
Conclusion: It seems that the coordination between three main factors, namely high dietary carbohydrate intake, insulin resistance, and hyperandrogenism, can be considered as the initiator for inflammation induction and oxidative stress in PCOS patients. The antioxidant supplements and anti-inflammatory agents can be useful to improve the results of infertility treatment cycles in these patients; however, concluding the results still requires further studies.

کلیدواژه‌ها [English]

  • Antioxidant
  • Assisted reproductive technique
  • Inflammation
  • Oxidative stress
  • Polycystic ovary syndrome
  1. Goldrat O, Delbaere A. PCOS: update and diagnostic approach. Clinical biochemistry 2018; 62:24-31.
  2. El Hayek S, Bitar L, Hamdar LH, Mirza FG, Daoud G. Poly cystic ovarian syndrome: an updated overview. Frontiers in physiology 2016; 7:124.
  3. Kalhori Z, Mehranjani MS, Azadbakht M, Shariatzadeh MA. L-Carnitine improves endocrine function and folliculogenesis by reducing inflammation, oxidative stress and apoptosis in mice following induction of polycystic ovary syndrome. Reproduction, Fertility and Development 2019; 31(2):282-93.
  4. Wang R, Mol BW. The Rotterdam criteria for polycystic ovary syndrome: evidence-based criteria?. Human Reproduction 2017; 32(2):261-4.
  5. Morgante G, Massaro MG, Di Sabatino A, Cappelli V, De Leo V. Therapeutic approach for metabolic disorders and infertility in women with PCOS. Gynecological Endocrinology 2018; 34(1):4-9.
  6. Kelly CC, Lyall H, Petrie JR, Gould GW, Connell JM, Sattar N. Low grade chronic inflammation in women with polycystic ovarian syndrome. The Journal of Clinical Endocrinology & Metabolism 2001; 86(6):2453-5.
  7. Patel S. Polycystic ovary syndrome (PCOS), an inflammatory, systemic, lifestyle endocrinopathy. J Steroid Biochem Mol Biol 2018; 182:27-36.
  8. Hong L, Zhang Y, Wang Q, Han Y, Teng X. Effects of interleukin 6 and tumor necrosis factor-α on the proliferation of porcine theca interna cells: Possible role of these cytokines in the pathogenesis of polycystic ovary syndrome. Taiwan J Obstet Gynecol 2016; 55(2):183-7.
  9. Furat Rencber S, Kurnaz Ozbek S, Eraldemır 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(1):1-6.
  10. Brouillet S, Boursier G, Anav M, Du Boulet De La Boissière B, Gala A, Ferrieres-Hoa A, et al. C-reactive protein and ART outcomes: a systematic review. Human Reproduction Update 2020; 26(5):753-73.
  11. Kahyaoglu S, Yumuşak OH, Ozyer S, Pekcan MK, Erel M, Cicek MN, et al. Clomiphene citrate treatment cycle outcomes of polycystic ovary syndrome patients based on basal high sensitive C-reactive protein levels: a cross-sectional study. International Journal of Fertility & Sterility 2017; 10(4):320.
  12. Escobar-Morreale HF, Luque-Ramírez M, González F. Circulating inflammatory markers in polycystic ovary syndrome: a systematic review and metaanalysis. Fertil Steril 2011; 95(3):1048-58.
  13. Borthakur A, Prabhu YD, Gopalakrishnan AV. Role of IL-6 signalling in polycystic ovarian syndrome associated inflammation. Journal of Reproductive Immunology 2020; 141:103155.
  14. Peng Z, Sun Y, Lv X, Zhang H, Liu C, Dai S. Interleukin-6 levels in women with polycystic ovary syndrome: a systematic review and meta-analysis. PloS one 2016; 11(2):e0148531.
  15. Gao L, Gu Y, Yin X. High serum tumor necrosis factor-alpha levels in women with polycystic ovary syndrome: a meta-analysis. PloS one 2016; 11(10):e0164021.
  16. Stephens JM, Pekala PH. Transcriptional repression of the GLUT4 and C/EBP genes in 3T3-L1 adipocytes by tumor necrosis factor-alpha. Journal of Biological Chemistry 1991; 266(32):21839-45.
  17. Amato G, Conte M, Mazziotti G, Lalli E, Vitolo G, Tucker AT, et al. Serum and follicular fluid cytokines in polycystic ovary syndrome during stimulated cycles. Obstetrics & Gynecology 2003; 101(6):1177-82.
  18. Rostamtabar M, Esmaeilzadeh S, Tourani M, Rahmani A, Baee M, Shirafkan F, et al. Pathophysiological roles of chronic low‐grade inflammation mediators in polycystic ovary syndrome. Journal of Cellular Physiology 2021; 236(2):824-38.
  19. Yang Y, Qiao J, Li R, Li MZ. Is interleukin-18 associated with polycystic ovary syndrome?. Reprod Biol Endocrinol 2011; 9(1):1-5.
  20. Günther V, Alkatout I, Fuhs C, Salmassi A, Mettler L, Hedderich J, et al. The role of interleukin-18 in serum and follicular fluid during in vitro fertilization and intracytoplasmic sperm injection. BioMed research international 2016; 2016.
  21. Matsumoto T, Kobayashi T, Kamata K. Relationships among ET-1, PPARγ, oxidative stress and endothelial dysfunction in diabetic animals. Journal of Smooth Muscle Research 2008; 44(2):41-55.
  22. Koleva DI, Orbetzova MM, Nikolova JG, Tyutyundzhiev SB. Adipokines and soluble cell adhesion molecules in insulin resistant and non-insulin resistant women with polycystic ovary syndrome. Archives of Physiology and Biochemistry 2016; 122(4):223-7.
  23. Solano ME, Sander VA, Ho H, Motta AB, Arck PC. Systemic inflammation, cellular influx and up-regulation of ovarian VCAM-1 expression in a mouse model of polycystic ovary syndrome (PCOS). Journal of Reproductive Immunology 2011; 92(1-2):33-44.
  24. Holecki M, Zahorska‐Markiewicz B, Janowska J, Nieszporek T, Wojaczyńska‐Stanek K, Żak‐Gołąb A, et al. The influence of weight loss on serum osteoprotegerin concentration in obese perimenopausal women. Obesity 2007; 15(8):1925-9.
  25. Yang A, Pizzulli L, Lüderitz B. Mean platelet volume as marker of restenosis after percutaneous transluminal coronary angioplasty in patients with stable and unstable angina pectoris. Thromb Res 2006; 117(4):371-7.
  26. Diamanti-Kandarakis E. Polycystic ovarian syndrome: pathophysiology, molecular aspects and clinical implications. Expert Reviews in molecular medicine 2008; 10.
  27. Luque-Ramírez M, Álvarez-Blasco F, Botella-Carretero JI, Sanchón R, San Millán JL, Escobar-Morreale HF. Increased body iron stores of obese women with polycystic ovary syndrome are a consequence of insulin resistance and hyperinsulinism and are not a result of reduced menstrual losses. Diabetes Care 2007; 30(9):2309-13.
  28. Donkena KV, Young CY, Tindall DJ. Oxidative stress and DNA methylation in prostate cancer. Obstet Gynecol Int 2010; 2010.
  29. Franco R, Schoneveld O, Georgakilas AG, Panayiotidis MI. Oxidative stress, DNA methylation and carcinogenesis. Cancer Lett 2008; 266(1):6-11.
  30. Touyz RM. Molecular and cellular mechanisms in vascular injury in hypertension: role of angiotensin II–editorial review. C Curr Opin Nephrol Hypertens 2005; 14(2):125-31.
  31. Siti HN, Kamisah Y, Kamsiah JJ. The role of oxidative stress, antioxidants and vascular inflammation in cardiovascular disease (a review). Vascul Pharmacol 2015; 71:40-56.
  32. Zuo T, Zhu M, Xu W. Roles of oxidative stress in polycystic ovary syndrome and cancers. Oxid Med Cell Longev 2016; 2016.
  33. Nasiri N, Moini A, Eftekhari-Yazdi P, Karimian L, Salman-Yazdi R, Zolfaghari Z, et al. Abdominal obesity can induce both systemic and follicular fluid oxidative stress independent from polycystic ovary syndrome. Eur J Obstet Gynecol Reprod Biol 2015; 184:112-6.
  34. Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked?. F Free Radic Biol Med 2010; 49(11):1603-16.
  35. Dumesic DA, Lobo RA. Cancer risk and PCOS. Steroids 2013; 78(8):782-5.
  36. Khashchenko E, Vysokikh M, Uvarova E, Krechetova L, Vtorushina V, Ivanets T, et al. Activation of Systemic Inflammation and Oxidative Stress in Adolescent Girls with Polycystic Ovary Syndrome in Combination with Metabolic Disorders and Excessive Body Weight. J Clin Med 2020; 9(5):1399.
  37. Melo AS, Kliemchen J, Junior AA, Ferriani RA, Navarro PA. Oxidative stress and polycystic ovary syndrome: evaluation during ovarian stimulation for ICSI. Reproduction (Cambridge, England); 2016.
  38. March WA, Moore VM, Willson KJ, Phillips DI, Norman RJ, Davies MJ. The prevalence of polycystic ovary syndrome in a community sample assessed under contrasting diagnostic criteria. Hum Reprod 2010; 25(2):544-51.
  39. Choi HD, Kim JH, Chang MJ, Kyu‐Youn Y, Shin WG. Effects of astaxanthin on oxidative stress in overweight and obese adults. Phytother Res 2011; 25(12):1813-8.
  40. Glintborg D. Endocrine and metabolic characteristics in polycystic ovary syndrome. Dan Med J 2016; 63(4).
  41. Ghaffarzad A, Amani R, Sadaghiani MM, Darabi M, Cheraghian B. Correlation of serum lipoprotein ratios with insulin resistance in infertile women with polycystic ovarian syndrome: A Case Control Study. Int J Fertil Steril 2016; 10(1):29.
  42. Imran A, Butt MS, Arshad MS, Arshad MU, Saeed F, Sohaib M, et al. Exploring the potential of black tea based flavonoids against hyperlipidemia related disorders. Lipids Health Dis 2018; 17(1):1-6.
  43. Couillard C, Ruel G, Archer WR, Pomerleau S, Bergeron J, Couture P, et al. Circulating levels of oxidative stress markers and endothelial adhesion molecules in men with abdominal obesity. J Clin Endocrinol Metab 2005; 90(12):6454-9.
  44. Broughton DE, Moley KH. Obesity and female infertility: potential mediators of obesity's impact. Fertility and sterility 2017; 107(4):840-7.
  45. Murri M, Luque-Ramírez M, Insenser M, Ojeda-Ojeda M, Escobar-Morreale HF. Circulating markers of oxidative stress and polycystic ovary syndrome (PCOS): a systematic review and meta-analysis. Human reproduction update 2013; 19(3):268-88.
  46. Klop B, Elte JW, Castro Cabezas M. Dyslipidemia in obesity: mechanisms and potential targets. Nutrients 2013; 5(4):1218-40.
  47. Salvado L, Palomer X, Barroso E, Vázquez-Carrera M. Targeting endoplasmic reticulum stress in insulin resistance. Trends in Endocrinology & Metabolism 2015; 26(8):438-48.
  48. Mohamadin AM, Habib FA, Elahi TF. Serum paraoxonase 1 activity and oxidant/antioxidant status in Saudi women with polycystic ovary syndrome. Pathophysiology 2010; 17(3):189-96.
  49. Dias JP, Talbot S, Sénécal J, Carayon P, Couture R. Kinin B1 receptor enhances the oxidative stress in a rat model of insulin resistance: outcome in hypertension, allodynia and metabolic complications. PloS one 2010; 5(9):e12622.
  50. Castro MC, Massa ML, Arbeláez LG, Schinella G, Gagliardino JJ, Francini F. Fructose-induced inflammation, insulin resistance and oxidative stress: A liver pathological triad effectively disrupted by lipoic acid. Life sciences 2015; 137:1-6.
  51. Rudich AS, Kozlovsky N, Potashnik RU, Bashan NA. Oxidant stress reduces insulin responsiveness in 3T3-L1 adipocytes. American Journal of Physiology-Endocrinology And Metabolism 1997; 272(5):E935-40.
  52. González F. Inflammation in polycystic ovary syndrome: underpinning of insulin resistance and ovarian dysfunction. Steroids 2012; 77(4):300-5.
  53. Keane KN, Cruzat VF, Carlessi R, De Bittencourt PI, Newsholme P. Molecular events linking oxidative stress and inflammation to insulin resistance and β-cell dysfunction. Oxidative medicine and cellular longevity 2015; 2015.
  54. Pollak M. The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nature Reviews Cancer 2012; 12(3):159-69.
  55. 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. The Journal of Clinical Endocrinology & Metabolism 1986; 62(5):904-10.
  56. Nisenblat V, Norman RJ. Androgens and polycystic ovary syndrome. Current Opinion in Endocrinology, Diabetes and Obesity 2009; 16(3):224-31.
  57. González F, Minium J, Rote NS, Kirwan JP. Hyperglycemia alters tumor necrosis factor-α release from mononuclear cells in women with polycystic ovary syndrome. The Journal of Clinical Endocrinology & Metabolism 2005; 90(9):5336-42.
  58. Spaczynski RZ, Arici A, Duleba AJ. Tumor necrosis factor-α stimulates proliferation of rat ovarian theca-interstitial cells. biology of reproduction 1999; 61(4):993-8.
  59. González F, Rote NS, Minium J, Kirwan JP. 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):336-40.
  60. Nikolić M, Macut D, Djordjevic A, Veličković N, Nestorović N, Bursać B, et al. Possible involvement of glucocorticoids in 5α-dihydrotestosterone-induced PCOS-like metabolic disturbances in the rat visceral adipose tissue. Molecular and cellular endocrinology 2015; 399:22-31.
  61. Tepavčević S, Milutinović DV, Macut D, Stanišić J, Nikolić M, Božić-Antić I, et al. Cardiac nitric oxide synthases and Na+/K+-ATPase in the rat model of polycystic ovary syndrome induced by dihydrotestosterone. Experimental and Clinical Endocrinology & Diabetes 2015; 123(05):303-7.
  62. Zheng YH, Ding T, Ye DF, Liu H, Lai MH, Ma HX. Effect of low-frequency electroacupuncture intervention on oxidative stress and glucose metabolism in rats with polycystic ovary syndrome. Zhen ci yan jiu= Acupuncture Research 2015; 40(2):125-30.
  63. Turan V, Sezer ED, Zeybek B, Sendag F. Infertility and the presence of insulin resistance are associated with increased oxidative stress in young, non-obese Turkish women with polycystic ovary syndrome. Journal of pediatric and adolescent gynecology 2015; 28(2):119-23.
  64. Lai Q, Xiang W, Li Q, Zhang H, Li Y, Zhu G, et al. Oxidative stress in granulosa cells contributes to poor oocyte quality and IVF-ET outcomes in women with polycystic ovary syndrome. Frontiers of medicine 2018; 12(5):518-24.
  65. Jakimiuk AJ, Weitsman SR, Navab A, Magoffin DA. Luteinizing hormone receptor, steroidogenesis acute regulatory protein, and steroidogenic enzyme messenger ribonucleic acids are overexpressed in thecal and granulosa cells from polycystic ovaries. The Journal of Clinical Endocrinology & Metabolism 2001; 86(3):1318-23.
  66. Pellatt L, Hanna L, Brincat M, Galea R, Brain H, Whitehead S, et al. Granulosa cell production of anti-Mullerian hormone is increased in polycystic ovaries. The Journal of Clinical Endocrinology & Metabolism 2007; 92(1):240-5.
  67. Jančar N, Kopitar AN, Ihan A, Klun IV, Bokal EV. Effect of apoptosis and reactive oxygen species production in human granulosa cells on oocyte fertilization and blastocyst development. Journal of assisted reproduction and genetics 2007; 24(2):91-7.
  68. Wassarman PM, Litscher ES. Influence of the zona pellucida of the mouse egg on folliculogenesis and fertility. International Journal of Developmental Biology 2013; 56(10-12):833-9.
  69. Balen AH, Tan SL, MacDougall J, Jacobs HS. Miscarriage rates following in-vitro fertilization are increased in women with polycystic ovaries and reduced by pituitary desensitization with buserelin. Human Reproduction 1993; 8(6):959-64.
  70. Morais RD, Thomé RG, Lemos FS, Bazzoli N, Rizzo E. Autophagy and apoptosis interplay during follicular atresia in fish ovary: a morphological and immunocytochemical study. Cell and tissue research 2012; 347(2):467-78.
  71. Xiong YL, Liang XY, Yang X, Li Y, Wei LN. Low-grade chronic inflammation in the peripheral blood and ovaries of women with polycystic ovarian syndrome. European Journal of Obstetrics & Gynecology and Reproductive Biology 2011; 159(1):148-50.
  72. Yang F, Ruan YC, Yang YJ, Wang K, Liang SS, Han YB, et al. Follicular hyperandrogenism downregulates aromatase in luteinized granulosa cells in polycystic ovary syndrome women. Reproduction 2015; 150(4):289-96.
  73. Balen AH. Hypersecretion of luteinizing hormone in the polycystic ovary syndrome and a novel hormone'gonadotrophin surge attenuating factor'. Journal of the Royal Society of Medicine 1995; 88(6):339P.
  74. Agarwal A, Aponte-Mellado A, Premkumar BJ, Shaman A, Gupta S. The effects of oxidative stress on female reproduction: a review. Reproductive biology and endocrinology 2012; 10(1):1-31.
  75. Fabjan T, Bokal EV, Klun IV, Bedenk J, Kumer K, Osredkar J. Antimüllerian Hormone and Oxidative Stress Biomarkers as Predictors of Successful Preg-nancy in Polycystic Ovary Syndrome, Endometriosis and Tubal Infertility Factor. Acta Chimica Slovenica 2020; 67(3):885-95.
  76. Liu Y, Yu Z, Zhao S, Cheng L, Man Y, Gao X, et al. Oxidative stress markers in the follicular fluid of patients with polycystic ovary syndrome correlate with a decrease in embryo quality. Journal of Assisted Reproduction and Genetics 2021; 38(2):471-7.
  77. Banaszewska B, Pawelczyk L, Spaczynski R. Current and future aspects of several adjunctive treatment strategies in polycystic ovary syndrome. Reproductive biology 2019; 19(4):309-15.
  78. Artini PG, Obino ME, Sergiampietri C, Pinelli S, Papini F, Casarosa E, et al. PCOS and pregnancy: a review of available therapies to improve the outcome of pregnancy in women with polycystic ovary syndrome. Expert review of endocrinology & metabolism 2018; 13(2):87-98.
  79. Mojaverrostami S, Asghari N, Khamisabadi M, Khoei HH. The role of melatonin in polycystic ovary syndrome: A review. International Journal of Reproductive BioMedicine 2019; 17(12):865.
  80. Jain P, Jain M, Haldar C, Singh TB, Jain S. Melatonin and its correlation with testosterone in polycystic ovarian syndrome. Journal of human reproductive sciences 2013; 6(4):253.
  81. Tamura H, Nakamura Y, Korkmaz A, Manchester LC, Tan DX, Sugino N, et al. Melatonin and the ovary: physiological and pathophysiological implications. Fertility and sterility 2009; 92(1):328-43.
  82. Javanmanesh F, Kashanian M, Rahimi M, Sheikhansari N. A comparison between the effects of metformin and N-acetyl cysteine (NAC) on some metabolic and endocrine characteristics of women with polycystic ovary syndrome. Gynecological Endocrinology 2016; 32(4):285-9.
  83. Abdelrazik H, Agarwal A. L-carnitine and assisted reproduction. Archives of Medical Science Special Issues 2009; 5:43-7.
  84. Kitano Y, Hashimoto S, Matsumoto H, Yamochi T, Yamanaka M, Nakaoka Y, et al. Oral administration of l-carnitine improves the clinical outcome of fertility in patients with IVF treatment. Gynecological Endocrinology 2018; 34(8):684-8.
  85. Florou P, Anagnostis P, Theocharis P, Chourdakis M, Goulis DG. Does coenzyme Q10 supplementation improve fertility outcomes in women undergoing assisted reproductive technology procedures? A systematic review and meta-analysis of randomized-controlled trials. Journal of assisted reproduction and genetics 2020; 37(10):2377-87.
  86. Al-Qadhi HI, Kadhim EJ, Ali RH. Coenzyme Q10 effects on body weight, serum testosterone level and oxidative stress in women with polycystic ovarian syndrome (PCOS). International Journal of Research in Pharmaceutical Sciences 2017; 8(3):377-82.
  87. Agarwal A, Durairajanayagam D, Du Plessis SS. Utility of antioxidants during assisted reproductive techniques: an evidence based review. Reproductive Biology and Endocrinology 2014; 12(1):1-9.
  88. Kurdoglu Z, Ozkol H, Tuluce Y, Koyuncu I. Oxidative status and its relation with insulin resistance in young non-obese women with polycystic ovary syndrome. Journal of Endocrinological Investigation 2012; 35(3):317-21.
  89. Teegarden D, Donkin SS. Vitamin D: emerging new roles in insulin sensitivity. Nutrition research reviews 2009; 22(1):82-92.
  90. Zoidis E, Seremelis I, Kontopoulos N, Danezis GP. Selenium-dependent antioxidant enzymes: Actions and properties of selenoproteins. Antioxidants 2018; 7(5):66.
  91. Wang Z, Zhai D, Zhang D, Bai L, Yao R, Yu J, et al. Quercetin decreases insulin resistance in a polycystic ovary syndrome rat model by improving inflammatory microenvironment. Reproductive Sciences 2017; 24(5):682-90.
  92. Lakhanpal P, Rai DK. Quercetin: a versatile flavonoid. Internet Journal of Medical Update 2007; 2(2):22-37.
  93. Hasanpour Dehkordi A, Abbaszadeh A, Mir S, Hasanvand A. Metformin and its anti-inflammatory and anti-oxidative effects; new concepts. Journal of Renal Injury Prevention; 2019.
  94. An H, He L. Current understanding of metformin effect on the control of hyperglycemia in diabetes. The Journal of endocrinology 2016; 228(3):R97.
  95. Della Corte L, Foreste V, Barra F, Gustavino C, Alessandri F, Centurioni MG, et al. Current and experimental drug therapy for the treatment of polycystic ovarian syndrome. Expert Opinion on Investigational Drugs 2020; 29(8):819-30.
  96. Oner G, Muderris II. Efficacy of omega-3 in the treatment of polycystic ovary syndrome. Journal of Obstetrics and Gynaecology 2013; 33(3):289-91.
  97. Rashidi B, Abediasl J, Tehraninejad E, Rahmanpour H, Sills ES. Simvastatin effects on androgens, inflammatory mediators, and endogenous pituitary gonadotropins among patients with PCOS undergoing IVF: results from a prospective, randomized, placebo-controlled clinical trial. Journal of Investigative Medicine 2011; 59(6):912-6.
  98. Ramírez-Garza SL, Laveriano-Santos EP, Marhuenda-Muñoz M, Storniolo CE, Tresserra-Rimbau A, Vallverdú-Queralt A, et al. Health effects of resveratrol: Results from human intervention trials. Nutrients 2018; 10(12):1892.