Skip to main content
Top
Published in: Journal of Ovarian Research 1/2020

01-12-2020 | Insulins | Review

Quercetin and polycystic ovary syndrome, current evidence and future directions: a systematic review

Authors: Fatemeh Pourteymour Fard Tabrizi, Fatemeh Hajizadeh-Sharafabad, Maryam Vaezi, Hamed Jafari-Vayghan, Mohammad Alizadeh, Vahid Maleki

Published in: Journal of Ovarian Research | Issue 1/2020

Login to get access

Abstract

Polycystic ovary syndrome (PCOS) is a polygenic endocrine disorder and the most common gynecological endocrinopathy among reproductive-aged women. Current remedies are often used only to control its signs and symptoms, while they are not thoroughly able to prevent complications. Quercetin is an herbal bioactive flavonoid commonly used for the treatment of metabolic and inflammatory disorders. Thus, this systematic review was conducted to evaluate the efficacy of quercetin supplementation in subjects with PCOS. Databases until March 2019 were searched. All human clinical trials and animal models evaluating the effects of quercetin on PCOS women were included. Out of 253 articles identified in our search, 8 eligible articles (5 animal studies and 3 clinical trials) were reviewed. The majority of studies supported the beneficial effects of quercetin on the ovarian histomorphology, folliculogenesis, and luteinisation processes. The effects of quercetin on reducing the levels of testosterone, luteinizing hormone (LH), and insulin resistance were also reported. Although quercetin improved dyslipidemia, no significant effect was reported for weight loss. It is suggested that the benefits of quercetin may be more closely related to antioxidant and anti-inflammatory features of quercetin rather than weight-reducing effects. Therefore, this review article provides evidence that quercetin could be considered as a potential agent to attenuate PCOS complications. However, due to the paucity of high-quality clinical trials, further studies are needed.
Literature
1.
go back to reference Ding T, Hardiman PJ, Petersen I, Wang F-F, Qu F, Baio G. The prevalence of polycystic ovary syndrome in reproductive-aged women of different ethnicity: a systematic review and meta-analysis. Oncotarget. 2017;8:96351.PubMedPubMedCentralCrossRef Ding T, Hardiman PJ, Petersen I, Wang F-F, Qu F, Baio G. The prevalence of polycystic ovary syndrome in reproductive-aged women of different ethnicity: a systematic review and meta-analysis. Oncotarget. 2017;8:96351.PubMedPubMedCentralCrossRef
2.
go back to reference Sirmans SM, Pate KA. Epidemiology, diagnosis, and management of polycystic ovary syndrome. Clin Epidemiol. 2014;6:1. Sirmans SM, Pate KA. Epidemiology, diagnosis, and management of polycystic ovary syndrome. Clin Epidemiol. 2014;6:1.
3.
go back to reference Blay SL, Aguiar JVA, Passos IC. Polycystic ovary syndrome and mental disorders: a systematic review and exploratory meta-analysis. Neuropsychiatr Dis Treat. 2016;12:2895.PubMedPubMedCentralCrossRef Blay SL, Aguiar JVA, Passos IC. Polycystic ovary syndrome and mental disorders: a systematic review and exploratory meta-analysis. Neuropsychiatr Dis Treat. 2016;12:2895.PubMedPubMedCentralCrossRef
4.
go back to reference Essah PA, Nestler JE. The metabolic syndrome in polycystic ovary syndrome. J Endocrinol Invest. 2006;29:270–80.PubMedCrossRef Essah PA, Nestler JE. The metabolic syndrome in polycystic ovary syndrome. J Endocrinol Invest. 2006;29:270–80.PubMedCrossRef
5.
go back to reference Beydoun HA, Stadtmauer L, Beydoun MA, Russell H, Zhao Y, Oehninger S. Polycystic ovary syndrome, body mass index and outcomes of assisted reproductive technologies. Reprod Biomed Online. 2009;18:856–63.PubMedPubMedCentralCrossRef Beydoun HA, Stadtmauer L, Beydoun MA, Russell H, Zhao Y, Oehninger S. Polycystic ovary syndrome, body mass index and outcomes of assisted reproductive technologies. Reprod Biomed Online. 2009;18:856–63.PubMedPubMedCentralCrossRef
6.
go back to reference Orio F, Muscogiuri G, Nese C, Palomba S, Savastano S, Tafuri D, et al. Obesity, type 2 diabetes mellitus and cardiovascular disease risk: an uptodate in the management of polycystic ovary syndrome. Eur J Obstet Gynecol Reprod Biol. 2016;207:214–9.PubMedCrossRef Orio F, Muscogiuri G, Nese C, Palomba S, Savastano S, Tafuri D, et al. Obesity, type 2 diabetes mellitus and cardiovascular disease risk: an uptodate in the management of polycystic ovary syndrome. Eur J Obstet Gynecol Reprod Biol. 2016;207:214–9.PubMedCrossRef
7.
go back to reference McEwen B, Hartmann G. Insulin resistance’and polycystic ovary syndrome (PCOS)’: part 1. The impact of insulin resistance. J Aust Tradit Soc. 2018;24:214. McEwen B, Hartmann G. Insulin resistance’and polycystic ovary syndrome (PCOS)’: part 1. The impact of insulin resistance. J Aust Tradit Soc. 2018;24:214.
8.
go back to reference Legro RS, Kunselman AR, Dodson WC, Dunaif A. Prevalence and predictors of risk for type 2 diabetes mellitus and impaired glucose tolerance in polycystic ovary syndrome: a prospective, controlled study in 254 affected women. J Clin Endocrinol Metab. 1999;84:165–9.PubMed Legro RS, Kunselman AR, Dodson WC, Dunaif A. Prevalence and predictors of risk for type 2 diabetes mellitus and impaired glucose tolerance in polycystic ovary syndrome: a prospective, controlled study in 254 affected women. J Clin Endocrinol Metab. 1999;84:165–9.PubMed
9.
go back to reference Shukla A, Mandel L. Polycystic ovarian syndrome. Obes Manag. 2019:1:31–40. Shukla A, Mandel L. Polycystic ovarian syndrome. Obes Manag. 2019:1:31–40.
10.
go back to reference Kakoly NS, Earnest A, Teede HJ, Moran LJ, Joham AE. The impact of obesity on the incidence of type 2 diabetes among women with polycystic ovary syndrome. Diabetes Care. 2019;42:560–7.PubMedCrossRef Kakoly NS, Earnest A, Teede HJ, Moran LJ, Joham AE. The impact of obesity on the incidence of type 2 diabetes among women with polycystic ovary syndrome. Diabetes Care. 2019;42:560–7.PubMedCrossRef
11.
go back to reference Rocca ML, Venturella R, Mocciaro R, Di Cello A, Sacchinelli A, Russo V, et al. Polycystic ovary syndrome: chemical pharmacotherapy. Expert Opin Pharmacother. 2015;16:1369–93.PubMedCrossRef Rocca ML, Venturella R, Mocciaro R, Di Cello A, Sacchinelli A, Russo V, et al. Polycystic ovary syndrome: chemical pharmacotherapy. Expert Opin Pharmacother. 2015;16:1369–93.PubMedCrossRef
12.
go back to reference David AVA, Arulmoli R, Parasuraman S. Overviews of biological importance of quercetin: a bioactive flavonoid. Pharmacogn Rev. 2016;10:84.CrossRef David AVA, Arulmoli R, Parasuraman S. Overviews of biological importance of quercetin: a bioactive flavonoid. Pharmacogn Rev. 2016;10:84.CrossRef
13.
go back to reference Lakhanpal P, Rai DK. Quercetin: a versatile flavonoid. Internet J Med Updat. 2007;2:22–37. Lakhanpal P, Rai DK. Quercetin: a versatile flavonoid. Internet J Med Updat. 2007;2:22–37.
14.
go back to reference Boots AW, Haenen GRMM, Bast A. Health effects of quercetin: from antioxidant to nutraceutical. Eur J Pharmacol. 2008;585:325–37.PubMedCrossRef Boots AW, Haenen GRMM, Bast A. Health effects of quercetin: from antioxidant to nutraceutical. Eur J Pharmacol. 2008;585:325–37.PubMedCrossRef
15.
go back to reference Lesjak M, Beara I, Simin N, Pintać D, Majkić T, Bekvalac K, et al. Antioxidant and anti-inflammatory activities of quercetin and its derivatives. J Funct Foods. 2018;40:68–75.CrossRef Lesjak M, Beara I, Simin N, Pintać D, Majkić T, Bekvalac K, et al. Antioxidant and anti-inflammatory activities of quercetin and its derivatives. J Funct Foods. 2018;40:68–75.CrossRef
17.
go back to reference Gormaz JG, Quintremil S, Rodrigo R. Cardiovascular disease: a target for the pharmacological effects of quercetin. Curr Top Med Chem. 2015;15:1735–42.PubMedCrossRef Gormaz JG, Quintremil S, Rodrigo R. Cardiovascular disease: a target for the pharmacological effects of quercetin. Curr Top Med Chem. 2015;15:1735–42.PubMedCrossRef
18.
go back to reference Faggio C, Sureda A, Morabito S, Sanches-Silva A, Mocan A, Nabavi SF, et al. Flavonoids and platelet aggregation: a brief review. Eur J Pharmacol. 2017;807:91–101.PubMedCrossRef Faggio C, Sureda A, Morabito S, Sanches-Silva A, Mocan A, Nabavi SF, et al. Flavonoids and platelet aggregation: a brief review. Eur J Pharmacol. 2017;807:91–101.PubMedCrossRef
19.
go back to reference Shen Y, Croft KD, Hodgson JM, Kyle R, Lee I-LE, Wang Y, et al. Quercetin and its metabolites improve vessel function by inducing eNOS activity via phosphorylation of AMPK. Biochem Pharmacol. 2012;84:1036–44.PubMedCrossRef Shen Y, Croft KD, Hodgson JM, Kyle R, Lee I-LE, Wang Y, et al. Quercetin and its metabolites improve vessel function by inducing eNOS activity via phosphorylation of AMPK. Biochem Pharmacol. 2012;84:1036–44.PubMedCrossRef
20.
go back to reference Bondonno NP, Bondonno CP, Hodgson JM, Ward NC, Croft KD. The efficacy of quercetin in cardiovascular health. Curr Nutr Rep. 2015;4:290–303.CrossRef Bondonno NP, Bondonno CP, Hodgson JM, Ward NC, Croft KD. The efficacy of quercetin in cardiovascular health. Curr Nutr Rep. 2015;4:290–303.CrossRef
21.
go back to reference Perez-Vizcaino F, Duarte J, Jimenez R, Santos-Buelga C, Osuna A. Antihypertensive effects of the flavonoid quercetin. Pharmacol Rep. 2009;61:67–75.PubMedCrossRef Perez-Vizcaino F, Duarte J, Jimenez R, Santos-Buelga C, Osuna A. Antihypertensive effects of the flavonoid quercetin. Pharmacol Rep. 2009;61:67–75.PubMedCrossRef
22.
go back to reference Baghel SS, Shrivastava N, Baghel RS, Agrawal P, Rajput S. A review of quercetin: antioxidant and anticancer properties. World J Pharm Pharm Sci. 2012;1:146–60. Baghel SS, Shrivastava N, Baghel RS, Agrawal P, Rajput S. A review of quercetin: antioxidant and anticancer properties. World J Pharm Pharm Sci. 2012;1:146–60.
23.
go back to reference Rauf A, Imran M, Khan IA, ur-Rehman M, Gilani SA, Mehmood Z, et al. Anticancer potential of quercetin: a comprehensive review. Phyther Res. 2018;32:2109–30.CrossRef Rauf A, Imran M, Khan IA, ur-Rehman M, Gilani SA, Mehmood Z, et al. Anticancer potential of quercetin: a comprehensive review. Phyther Res. 2018;32:2109–30.CrossRef
24.
go back to reference Shi G-J, Li Y, Cao Q-H, Wu H-X, Tang X-Y, Gao X-H, et al. In vitro and in vivo evidence that quercetin protects against diabetes and its complications: a systematic review of the literature. Biomed Pharmacother. 2019;109:1085–99.PubMedCrossRef Shi G-J, Li Y, Cao Q-H, Wu H-X, Tang X-Y, Gao X-H, et al. In vitro and in vivo evidence that quercetin protects against diabetes and its complications: a systematic review of the literature. Biomed Pharmacother. 2019;109:1085–99.PubMedCrossRef
25.
go back to reference Eid HM, Haddad PS. The antidiabetic potential of quercetin: underlying mechanisms. Curr Med Chem. 2017;24:355–64.PubMedCrossRef Eid HM, Haddad PS. The antidiabetic potential of quercetin: underlying mechanisms. Curr Med Chem. 2017;24:355–64.PubMedCrossRef
26.
go back to reference Gurav M, Bhise S, Warghade S. Effect of Quercetin on Beta cell regeneration. Asian J Pharm Pharmacol. 2018;4:214–21.CrossRef Gurav M, Bhise S, Warghade S. Effect of Quercetin on Beta cell regeneration. Asian J Pharm Pharmacol. 2018;4:214–21.CrossRef
27.
go back to reference 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. Hum Reprod Update. 2013;19:268–88.PubMedCrossRef 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. Hum Reprod Update. 2013;19:268–88.PubMedCrossRef
29.
go back to reference Bentz AB. A review of Quercetin: chemistry, Antioxident properties, and bioavailability. J Young Investig. 2017;10:1–15. Bentz AB. A review of Quercetin: chemistry, Antioxident properties, and bioavailability. J Young Investig. 2017;10:1–15.
30.
go back to reference 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. Reprod Sci. 2017;24:682–90.PubMedCrossRef 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. Reprod Sci. 2017;24:682–90.PubMedCrossRef
31.
go back to reference Jahan S, Abid A, Khalid S, Afsar T, Shaheen G, Almajwal A, et al. Therapeutic potentials of Quercetin in management of polycystic ovarian syndrome using Letrozole induced rat model: a histological and a biochemical study. J Ovarian Res. 2018;11:26.PubMedPubMedCentralCrossRef Jahan S, Abid A, Khalid S, Afsar T, Shaheen G, Almajwal A, et al. Therapeutic potentials of Quercetin in management of polycystic ovarian syndrome using Letrozole induced rat model: a histological and a biochemical study. J Ovarian Res. 2018;11:26.PubMedPubMedCentralCrossRef
32.
go back to reference Neisy A, Zal F, Seghatoleslam A, Alaee S. Amelioration by quercetin of insulin resistance and uterine GLUT4 and ERα gene expression in rats with polycystic ovary syndrome (PCOS). Reprod Fertil Dev. 2019;31:315–23.PubMedCrossRef Neisy A, Zal F, Seghatoleslam A, Alaee S. Amelioration by quercetin of insulin resistance and uterine GLUT4 and ERα gene expression in rats with polycystic ovary syndrome (PCOS). Reprod Fertil Dev. 2019;31:315–23.PubMedCrossRef
33.
go back to reference Shah KN, Patel SS. Phosphatidylinositide 3-kinase inhibition: a new potential target for the treatment of polycystic ovarian syndrome. Pharm Biol. 2016;54:975–83.PubMedCrossRef Shah KN, Patel SS. Phosphatidylinositide 3-kinase inhibition: a new potential target for the treatment of polycystic ovarian syndrome. Pharm Biol. 2016;54:975–83.PubMedCrossRef
34.
go back to reference Hong Y, Yin Y, Tan Y, Hong K, Jiang F, Wang Y. Effect of quercetin on biochemical parameters in letrozoleinduced polycystic ovary syndrome in rats. Trop J Pharm Res. 2018;17:1783–8.CrossRef Hong Y, Yin Y, Tan Y, Hong K, Jiang F, Wang Y. Effect of quercetin on biochemical parameters in letrozoleinduced polycystic ovary syndrome in rats. Trop J Pharm Res. 2018;17:1783–8.CrossRef
35.
go back to reference Rezvan N, Moini A, Janani L, Mohammad K, Saedisomeolia A, Nourbakhsh M, et al. Effects of quercetin on adiponectin-mediated insulin sensitivity in polycystic ovary syndrome: a randomized placebo-controlled double-blind clinical trial. Horm Metab Res. 2017;49:115–21.PubMed Rezvan N, Moini A, Janani L, Mohammad K, Saedisomeolia A, Nourbakhsh M, et al. Effects of quercetin on adiponectin-mediated insulin sensitivity in polycystic ovary syndrome: a randomized placebo-controlled double-blind clinical trial. Horm Metab Res. 2017;49:115–21.PubMed
36.
go back to reference Khorshidi M, Moini A, Alipoor E, Rezvan N, Gorgani-Firuzjaee S, Yaseri M, et al. The effects of quercetin supplementation on metabolic and hormonal parameters as well as plasma concentration and gene expression of resistin in overweight or obese women with polycystic ovary syndrome. Phyther Res. 2018;32:2282–9.CrossRef Khorshidi M, Moini A, Alipoor E, Rezvan N, Gorgani-Firuzjaee S, Yaseri M, et al. The effects of quercetin supplementation on metabolic and hormonal parameters as well as plasma concentration and gene expression of resistin in overweight or obese women with polycystic ovary syndrome. Phyther Res. 2018;32:2282–9.CrossRef
37.
go back to reference Rezvan N, Moini A, Gorgani-Firuzjaee S, Hosseinzadeh-Attar MJ. Oral quercetin supplementation enhances adiponectin receptor transcript expression in polycystic ovary syndrome patients: a randomized placebo-controlled double-blind clinical trial. Cell J. 2018;19:627.PubMed Rezvan N, Moini A, Gorgani-Firuzjaee S, Hosseinzadeh-Attar MJ. Oral quercetin supplementation enhances adiponectin receptor transcript expression in polycystic ovary syndrome patients: a randomized placebo-controlled double-blind clinical trial. Cell J. 2018;19:627.PubMed
38.
go back to reference Lu M, Tang Q, Olefsky JM, Mellon PL, Webster NJG. Adiponectin activates adenosine monophosphate-activated protein kinase and decreases luteinizing hormone secretion in LβT2 gonadotropes. Mol Endocrinol. 2008;22:760–71.PubMedCrossRef Lu M, Tang Q, Olefsky JM, Mellon PL, Webster NJG. Adiponectin activates adenosine monophosphate-activated protein kinase and decreases luteinizing hormone secretion in LβT2 gonadotropes. Mol Endocrinol. 2008;22:760–71.PubMedCrossRef
39.
go back to reference Kafali H, Iriadam M, Ozardalı I, Demir N. Letrozole-induced polycystic ovaries in the rat: a new model for cystic ovarian disease. Arch Med Res. 2004;35:103–8.PubMedCrossRef Kafali H, Iriadam M, Ozardalı I, Demir N. Letrozole-induced polycystic ovaries in the rat: a new model for cystic ovarian disease. Arch Med Res. 2004;35:103–8.PubMedCrossRef
40.
go back to reference Wang F, Yu B, Yang W, Liu J, Lu J, Xia X. Polycystic ovary syndrome resembling histopathological alterations in ovaries from prenatal androgenized female rats. J Ovarian Res. 2012;5:15.PubMedPubMedCentralCrossRef Wang F, Yu B, Yang W, Liu J, Lu J, Xia X. Polycystic ovary syndrome resembling histopathological alterations in ovaries from prenatal androgenized female rats. J Ovarian Res. 2012;5:15.PubMedPubMedCentralCrossRef
41.
go back to reference Rezvanfar MA, Rezvanfar MA, Ahmadi A, Saadi HAS, Baeeri M, Abdollahi M. Mechanistic links between oxidative/nitrosative stress and tumor necrosis factor alpha in letrozole-induced murine polycystic ovary: biochemical and pathological evidences for beneficial effect of pioglitazone. Hum Exp Toxicol. 2012;31:887–97.PubMedCrossRef Rezvanfar MA, Rezvanfar MA, Ahmadi A, Saadi HAS, Baeeri M, Abdollahi M. Mechanistic links between oxidative/nitrosative stress and tumor necrosis factor alpha in letrozole-induced murine polycystic ovary: biochemical and pathological evidences for beneficial effect of pioglitazone. Hum Exp Toxicol. 2012;31:887–97.PubMedCrossRef
42.
go back to reference Doi SAR, Al-Zaid M, Towers PA, Scott CJ, Al-Shoumer KAS. Irregular cycles and steroid hormones in polycystic ovary syndrome. Hum Reprod. 2005;20:2402–8.PubMedCrossRef Doi SAR, Al-Zaid M, Towers PA, Scott CJ, Al-Shoumer KAS. Irregular cycles and steroid hormones in polycystic ovary syndrome. Hum Reprod. 2005;20:2402–8.PubMedCrossRef
43.
go back to reference Fukuda S, Orisaka M, Tajima K, Hattori K, Kotsuji F. Luteinizing hormone-induced Akt phosphorylation and androgen production are modulated by MAP kinase in bovine theca cells. J Ovarian Res. 2009;2:17.PubMedPubMedCentralCrossRef Fukuda S, Orisaka M, Tajima K, Hattori K, Kotsuji F. Luteinizing hormone-induced Akt phosphorylation and androgen production are modulated by MAP kinase in bovine theca cells. J Ovarian Res. 2009;2:17.PubMedPubMedCentralCrossRef
44.
go back to reference Singh A, Suragani M, Ehtesham NZ, Krishna A. Localization of resistin and its possible roles in the ovary of a vespertilionid bat, Scotophilus heathi. Steroids. 2015;95:17–23.PubMedCrossRef Singh A, Suragani M, Ehtesham NZ, Krishna A. Localization of resistin and its possible roles in the ovary of a vespertilionid bat, Scotophilus heathi. Steroids. 2015;95:17–23.PubMedCrossRef
45.
go back to reference Munir I, Yen H-W, Baruth T, Tarkowski R, Azziz R, Magoffin DA, et al. Resistin stimulation of 17α-hydroxylase activity in ovarian theca cells in vitro: relevance to polycystic ovary syndrome. J Clin Endocrinol Metab. 2005;90:4852–7.PubMedCrossRef Munir I, Yen H-W, Baruth T, Tarkowski R, Azziz R, Magoffin DA, et al. Resistin stimulation of 17α-hydroxylase activity in ovarian theca cells in vitro: relevance to polycystic ovary syndrome. J Clin Endocrinol Metab. 2005;90:4852–7.PubMedCrossRef
46.
go back to reference Rak-Mardyła A, Durak M, Gregoraszczuk EŁ. Effects of resistin on porcine ovarian follicle steroidogenesis in prepubertal animals: an in vitro study. Reprod Biol Endocrinol. 2013;11:45.PubMedPubMedCentralCrossRef Rak-Mardyła A, Durak M, Gregoraszczuk EŁ. Effects of resistin on porcine ovarian follicle steroidogenesis in prepubertal animals: an in vitro study. Reprod Biol Endocrinol. 2013;11:45.PubMedPubMedCentralCrossRef
47.
go back to reference Bremer AA, Miller WL. The serine phosphorylation hypothesis of polycystic ovary syndrome: a unifying mechanism for hyperandrogenemia and insulin resistance. Fertil Steril. 2008;89:1039–48.PubMedCrossRef Bremer AA, Miller WL. The serine phosphorylation hypothesis of polycystic ovary syndrome: a unifying mechanism for hyperandrogenemia and insulin resistance. Fertil Steril. 2008;89:1039–48.PubMedCrossRef
48.
go back to reference Nestler JE. Insulin resistance and the polycystic ovary syndrome: recent advances. Curr Opin Endocrinol Diabetes Obes. 2000;7:345–9.CrossRef Nestler JE. Insulin resistance and the polycystic ovary syndrome: recent advances. Curr Opin Endocrinol Diabetes Obes. 2000;7:345–9.CrossRef
49.
go back to reference 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. J Clin Endocrinol Metab. 2006;91:336–40.PubMedCrossRef 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. J Clin Endocrinol Metab. 2006;91:336–40.PubMedCrossRef
50.
go back to reference Franks S, Mason H, Willis D. Follicular dynamics in the polycystic ovary syndrome. Mol Cell Endocrinol. 2000;163:49–52.PubMedCrossRef Franks S, Mason H, Willis D. Follicular dynamics in the polycystic ovary syndrome. Mol Cell Endocrinol. 2000;163:49–52.PubMedCrossRef
51.
go back to reference Aguirre L, Arias N, Teresa Macarulla M, Gracia A, Portillo MP. Beneficial effects of quercetin on obesity and diabetes. Open Nutraceuticals J. 2011;4:189–198. Aguirre L, Arias N, Teresa Macarulla M, Gracia A, Portillo MP. Beneficial effects of quercetin on obesity and diabetes. Open Nutraceuticals J. 2011;4:189–198.
52.
go back to reference Pawlak K, Mysliwiec M, Pawlak D. Oxidized low-density lipoprotein (oxLDL) plasma levels and oxLDL to LDL ratio—are they real oxidative stress markers in dialyzed patients? Life Sci. 2013;92:253–8.PubMedCrossRef Pawlak K, Mysliwiec M, Pawlak D. Oxidized low-density lipoprotein (oxLDL) plasma levels and oxLDL to LDL ratio—are they real oxidative stress markers in dialyzed patients? Life Sci. 2013;92:253–8.PubMedCrossRef
53.
go back to reference Bhaskar S, Shalini V, Helen A. Quercetin regulates oxidized LDL induced inflammatory changes in human PBMCs by modulating the TLR-NF-κB signaling pathway. Immunobiology. 2011;216:367–73.PubMedCrossRef Bhaskar S, Shalini V, Helen A. Quercetin regulates oxidized LDL induced inflammatory changes in human PBMCs by modulating the TLR-NF-κB signaling pathway. Immunobiology. 2011;216:367–73.PubMedCrossRef
55.
go back to reference Mohamadin AM, Habib FA, Elahi TF. Serum paraoxonase 1 activity and oxidant/antioxidant status in Saudi women with polycystic ovary syndrome. Pathophysiology. 2010;17:189–96.PubMedCrossRef Mohamadin AM, Habib FA, Elahi TF. Serum paraoxonase 1 activity and oxidant/antioxidant status in Saudi women with polycystic ovary syndrome. Pathophysiology. 2010;17:189–96.PubMedCrossRef
56.
go back to reference Kadowaki T, Yamauchi T. Adiponectin receptor signaling: a new layer to the current model. Cell Metab. 2011;13:123–4.PubMedCrossRef Kadowaki T, Yamauchi T. Adiponectin receptor signaling: a new layer to the current model. Cell Metab. 2011;13:123–4.PubMedCrossRef
57.
go back to reference Satoh H, Nguyen MTA, Miles PDG, Imamura T, Usui I, Olefsky JM. Adenovirus-mediated chronic “hyper-resistinemia” leads to in vivo insulin resistance in normal rats. J Clin Invest. 2004;114:224–31.PubMedPubMedCentralCrossRef Satoh H, Nguyen MTA, Miles PDG, Imamura T, Usui I, Olefsky JM. Adenovirus-mediated chronic “hyper-resistinemia” leads to in vivo insulin resistance in normal rats. J Clin Invest. 2004;114:224–31.PubMedPubMedCentralCrossRef
58.
go back to reference Palanivel R, Maida A, Liu Y, Sweeney G. Regulation of insulin signalling, glucose uptake and metabolism in rat skeletal muscle cells upon prolonged exposure to resistin. Diabetologia. 2006;49:183–90.PubMedCrossRef Palanivel R, Maida A, Liu Y, Sweeney G. Regulation of insulin signalling, glucose uptake and metabolism in rat skeletal muscle cells upon prolonged exposure to resistin. Diabetologia. 2006;49:183–90.PubMedCrossRef
59.
go back to reference Seo JB, Noh MJ, Yoo EJ, Park SY, Park J, Lee IK, et al. Functional characterization of the human resistin promoter with adipocyte determination-and differentiation-dependent factor 1/sterol regulatory element binding protein 1c and CCAAT enhancer binding protein-α. Mol Endocrinol. 2003;17:1522–33.PubMedCrossRef Seo JB, Noh MJ, Yoo EJ, Park SY, Park J, Lee IK, et al. Functional characterization of the human resistin promoter with adipocyte determination-and differentiation-dependent factor 1/sterol regulatory element binding protein 1c and CCAAT enhancer binding protein-α. Mol Endocrinol. 2003;17:1522–33.PubMedCrossRef
60.
go back to reference Yang L, Li X, Gao L, Zhang Y, Cai G. Suppressive effects of quercetin-3-O-(6 ″-Feruloyl)-β-D-galactopyranoside on adipogenesis in 3T3-L1 preadipocytes through down-regulation of PPARγ and C/EBPα expression. Phyther Res. 2012;26:438–44.CrossRef Yang L, Li X, Gao L, Zhang Y, Cai G. Suppressive effects of quercetin-3-O-(6 ″-Feruloyl)-β-D-galactopyranoside on adipogenesis in 3T3-L1 preadipocytes through down-regulation of PPARγ and C/EBPα expression. Phyther Res. 2012;26:438–44.CrossRef
61.
go back to reference Lowes W, Walker M, Alberti KGMM, Agius L. Hexokinase isoenzymes in normal and cirrhotic human liver: suppression of glucokinase in cirrhosis. Biochim Biophys Acta (BBA)-General Subj. 1998;1379:134–42.CrossRef Lowes W, Walker M, Alberti KGMM, Agius L. Hexokinase isoenzymes in normal and cirrhotic human liver: suppression of glucokinase in cirrhosis. Biochim Biophys Acta (BBA)-General Subj. 1998;1379:134–42.CrossRef
62.
go back to reference Uno K, Katagiri H, Yamada T, Ishigaki Y, Ogihara T, Imai J, et al. Neuronal pathway from the liver modulates energy expenditure and systemic insulin sensitivity. Science (80- ). 2006;312:1656–9.CrossRef Uno K, Katagiri H, Yamada T, Ishigaki Y, Ogihara T, Imai J, et al. Neuronal pathway from the liver modulates energy expenditure and systemic insulin sensitivity. Science (80- ). 2006;312:1656–9.CrossRef
63.
go back to reference Liu M-L, Ylitalo K, Salonen R, Salonen JT, Taskinen M-R. Circulating oxidized low-density lipoprotein and its association with carotid intima-media thickness in asymptomatic members of familial combined hyperlipidemia families. Arterioscler Thromb Vasc Biol. 2004;24:1492–7.PubMedCrossRef Liu M-L, Ylitalo K, Salonen R, Salonen JT, Taskinen M-R. Circulating oxidized low-density lipoprotein and its association with carotid intima-media thickness in asymptomatic members of familial combined hyperlipidemia families. Arterioscler Thromb Vasc Biol. 2004;24:1492–7.PubMedCrossRef
Metadata
Title
Quercetin and polycystic ovary syndrome, current evidence and future directions: a systematic review
Authors
Fatemeh Pourteymour Fard Tabrizi
Fatemeh Hajizadeh-Sharafabad
Maryam Vaezi
Hamed Jafari-Vayghan
Mohammad Alizadeh
Vahid Maleki
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Journal of Ovarian Research / Issue 1/2020
Electronic ISSN: 1757-2215
DOI
https://doi.org/10.1186/s13048-020-0616-z

Other articles of this Issue 1/2020

Journal of Ovarian Research 1/2020 Go to the issue