The Implication of Mechanistic Approaches and the Role of the Microbiome in Polycystic Ovary Syndrome (PCOS): A Review
Abstract
:1. Introduction
2. PCOS
Transgenerational Transmission of PCOS and Its Pathophysiology
3. PCOS and COVID-19
4. Microbiome and PCOS
5. Gut Microbiomes and PCOS
5.1. Gut Microbiome Changes
5.2. IR concerning Gut Microbiomes
5.3. Sexual Hormones concerning the Gut Microbiome
5.4. Mechanism of Bile Acid and IL-22
5.5. SCFAs, Gut Hormones, and the Hypothalamic Pituitary Gonadal (HPG) Axis
5.6. Mechanism of Intestinal Permeability-LPS
6. Metabolites Contributing to the Development of PCOS
6.1. Microbiota Dysbiosis of PCOS
6.2. Bile Acids
6.3. Short-Chain Fatty Acids (SCFAs)
6.4. Ceramides
6.5. Trimethylamine N-Oxide (TMAO)
7. The Metabolic Perspective of PCOS
7.1. Interconnection between Insulin and PCOS
7.2. Obesity-Culprit in the Pathogenesis of PCOS
7.3. Inflammation
7.4. An Imbalance between Inflammatory Markers
7.4.1. Tumor Necrosis Factor-α (TNFα)
7.4.2. IL-6
7.4.3. IL-8
7.4.4. IL-10
7.4.5. IL-18
7.4.6. C-Reactive Protein (CRP)
8. Vaginal Microbiota
9. Metabolomic Insight into PCOS
10. MetS and PCOS
11. Serum Metabolomics in PCOS
Targeted and Untargeted Metabolomics
12. Metabolic Dysfunction in PCOS
12.1. IR in PCOS
12.2. Non-Alcoholic Fatty Liver Disease (NAFLD) and PCOS
12.3. Cardiovascular Disease (CVD) in PCOS
12.4. Other Metabolic Consequences of PCOS
13. A CLGI Process in PCOS
13.1. Metabolic and Inflammatory Markers
13.2. Pro-Inflammatory Cytokines and Chemokines
13.3. White Blood Cell Count (WBCs)
14. Therapeutic Opportunities
14.1. Probiotics, Prebiotics, and Synbiotics
14.2. Fecal Microbiota Transplantation
14.3. Short-Term Isoflavone Intervention in PCOS
14.4. Rhizomicrobiomics of Caesalpinia bonducella in PCOS Treatment
14.5. Metformin Treatment
14.6. Other Glucose-Lowering Medications
15. Conclusions
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Witchel, S.F.; Oberfield, S.E.; Peña, A.S. Polycystic Ovary Syndrome: Pathophysiology, Presentation, and Treatment with Emphasis on Adolescent Girls. J. Endocr. Soc. 2019, 3, 1545–1573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Legro, R.S.; Arslanian, S.A.; Ehrmann, D.A.; Hoeger, K.M.; Murad, M.H.; Pasquali, R.; Welt, C.K. Diagnosis and treatment of polycystic ovary syndrome: An Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2013, 98, 4565–4592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tabassum, F.; Jyoti, C.; Sinha, H.H.; Dhar, K.; Akhtar, M.S. Impact of polycystic ovary syndrome on quality of life of women in correlation to age, basal metabolic index, education and marriage. PLoS ONE 2021, 16, e0247486. [Google Scholar] [CrossRef] [PubMed]
- Brady, C.; Mousa, S.S.; Mousa, S.A. Polycystic ovary syndrome and its impact on women’s quality of life: More than just an endocrine disorder. Drug Healthc. Patient Saf. 2009, 1, 9–15. [Google Scholar] [CrossRef] [Green Version]
- He, F.-f.; Li, Y.-m. Role of gut microbiota in the development of insulin resistance and the mechanism underlying polycystic ovary syndrome: A review. J. Ovarian Res. 2020, 13, 73. [Google Scholar] [CrossRef]
- Wolf, W.M.; Wattick, R.A.; Kinkade, O.N.; Olfert, M.D. Geographical Prevalence of Polycystic Ovary Syndrome as Determined by Region and Race/Ethnicity. Int. J. Environ. Res. Public Health 2018, 15, 2589. [Google Scholar] [CrossRef] [Green Version]
- Rao, M.; Broughton, K.S.; LeMieux, M.J. Cross-sectional Study on the Knowledge and Prevalence of PCOS at a Multiethnic University. Prog. Prev. Med. 2020, e0028. [Google Scholar] [CrossRef]
- Kyrou, I.; Karteris, E.; Robbins, T.; Chatha, K.; Drenos, F.; Randeva, H.S. Polycystic ovary syndrome (PCOS) and COVID-19: An overlooked female patient population at potentially higher risk during the COVID-19 pandemic. BMC Med. 2020, 18, 220. [Google Scholar] [CrossRef]
- Rosenfield, R.L.; Ehrmann, D.A. The Pathogenesis of Polycystic Ovary Syndrome (PCOS): The Hypothesis of PCOS as Functional Ovarian Hyperandrogenism Revisited. Endocr. Rev. 2016, 37, 467–520. [Google Scholar] [CrossRef]
- Lie Fong, S.; Laven, J.S.E.; Duhamel, A.; Dewailly, D. Polycystic ovarian morphology and the diagnosis of polycystic ovary syndrome: Redefining threshold levels for follicle count and serum anti-Müllerian hormone using cluster analysis. Hum. Reprod. 2017, 32, 1723–1731. [Google Scholar] [CrossRef]
- Rao, P.; Bhide, P. Controversies in the diagnosis of polycystic ovary syndrome. Ther. Adv. Reprod. Health 2020, 14, 2633494120913032. [Google Scholar] [CrossRef]
- Reid, S.P.; Kao, C.-N.; Pasch, L.; Shinkai, K.; Cedars, M.I.; Huddleston, H.G. Ovarian morphology is associated with insulin resistance in women with polycystic ovary syndrome: A cross sectional study. Fertil. Res. Pract. 2017, 3, 8. [Google Scholar] [CrossRef] [Green Version]
- Teede, H.; Deeks, A.; Moran, L. Polycystic ovary syndrome: A complex condition with psychological, reproductive and metabolic manifestations that impacts on health across the lifespan. BMC Med. 2010, 8, 41. [Google Scholar] [CrossRef] [Green Version]
- Kelsey, T.W.; Dodwell, S.K.; Wilkinson, A.G.; Greve, T.; Andersen, C.Y.; Anderson, R.A.; Wallace, W.H. Ovarian volume throughout life: A validated normative model. PLoS ONE 2013, 8, e71465. [Google Scholar] [CrossRef] [Green Version]
- Johnstone, E.B.; Rosen, M.P.; Neril, R.; Trevithick, D.; Sternfeld, B.; Murphy, R.; Addauan-Andersen, C.; McConnell, D.; Pera, R.R.; Cedars, M.I. The polycystic ovary post-rotterdam: A common, age-dependent finding in ovulatory women without metabolic significance. J. Clin. Endocrinol. Metab. 2010, 95, 4965–4972. [Google Scholar] [CrossRef]
- Murphy, M.K.; Hall, J.E.; Adams, J.M.; Lee, H.; Welt, C.K. Polycystic ovarian morphology in normal women does not predict the development of polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 2006, 91, 3878–3884. [Google Scholar] [CrossRef]
- Alsamarai, S.; Adams, J.M.; Murphy, M.K.; Post, M.D.; Hayden, D.L.; Hall, J.E.; Welt, C.K. Criteria for polycystic ovarian morphology in polycystic ovary syndrome as a function of age. J. Clin. Endocrinol. Metab. 2009, 94, 4961–4970. [Google Scholar] [CrossRef] [Green Version]
- Merino, P.M.; Villarroel, C.; Jesam, C.; López, P.; Codner, E. New Diagnostic Criteria of Polycystic Ovarian Morphology for Adolescents: Impact on Prevalence and Hormonal Profile. Horm. Res. Paediatr. 2017, 88, 401–407. [Google Scholar] [CrossRef]
- Hart, R.; Doherty, D.A.; Norman, R.J.; Franks, S.; Dickinson, J.E.; Hickey, M.; Sloboda, D.M. Serum antimullerian hormone (AMH) levels are elevated in adolescent girls with polycystic ovaries and the polycystic ovarian syndrome (PCOS). Fertil. Steril. 2010, 94, 1118–1121. [Google Scholar] [CrossRef]
- Pigny, P.; Jonard, S.; Robert, Y.; Dewailly, D. Serum anti-Mullerian hormone as a surrogate for antral follicle count for definition of the polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 2006, 91, 941–945. [Google Scholar] [CrossRef]
- Rosenfield, R.L.; Wroblewski, K.; Padmanabhan, V.; Littlejohn, E.; Mortensen, M.; Ehrmann, D.A. Antimüllerian hormone levels are independently related to ovarian hyperandrogenism and polycystic ovaries. Fertil. Steril. 2012, 98, 242–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lizneva, D.; Suturina, L.; Walker, W.; Brakta, S.; Gavrilova-Jordan, L.; Azziz, R. Criteria, prevalence, and phenotypes of polycystic ovary syndrome. Fertil. Steril. 2016, 106, 6–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sachdeva, G.; Gainder, S.; Suri, V.; Sachdeva, N.; Chopra, S. Comparison of the Different PCOS Phenotypes Based on Clinical Metabolic, and Hormonal Profile, and their Response to Clomiphene. Indian J. Endocrinol. Metab. 2019, 23, 326–331. [Google Scholar] [CrossRef]
- Polak, A.M.; Adamska, A.; Krentowska, A.; Łebkowska, A.; Hryniewicka, J.; Adamski, M.; Kowalska, I. Body Composition, Serum Concentrations of Androgens and Insulin Resistance in Different Polycystic Ovary Syndrome Phenotypes. J. Clin. Med. 2020, 9, 732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mimouni, N.E.H.; Paiva, I.; Barbotin, A.L.; Timzoura, F.E.; Plassard, D.; Le Gras, S.; Ternier, G.; Pigny, P.; Catteau-Jonard, S.; Simon, V.; et al. Polycystic ovary syndrome is transmitted via a transgenerational epigenetic process. Cell Metab. 2021, 33, 513–530.e518. [Google Scholar] [CrossRef] [PubMed]
- Stener-Victorin, E.; Padmanabhan, V.; Walters, K.A.; Campbell, R.E.; Benrick, A.; Giacobini, P.; Dumesic, D.A.; Abbott, D.H. Animal Models to Understand the Etiology and Pathophysiology of Polycystic Ovary Syndrome. Endocr. Rev. 2020, 41, bnaa010. [Google Scholar] [CrossRef] [Green Version]
- Barrett, E.S.; Hoeger, K.M.; Sathyanarayana, S.; Abbott, D.H.; Redmon, J.B.; Nguyen, R.H.N.; Swan, S.H. Anogenital distance in newborn daughters of women with polycystic ovary syndrome indicates fetal testosterone exposure. J. Dev. Orig. Health Dis. 2018, 9, 307–314. [Google Scholar] [CrossRef]
- Dinsdale, N.L.; Crespi, B.J. Endometriosis and polycystic ovary syndrome are diametric disorders. Evol. Appl. 2021, 14, 1693–1715. [Google Scholar] [CrossRef]
- Bedenk, J.; Vrtačnik-Bokal, E.; Virant-Klun, I. The role of anti-Müllerian hormone (AMH) in ovarian disease and infertility. J. Assist. Reprod Genet. 2020, 37, 89–100. [Google Scholar] [CrossRef]
- Tata, B.; Mimouni, N.E.H.; Barbotin, A.L.; Malone, S.A.; Loyens, A.; Pigny, P.; Dewailly, D.; Catteau-Jonard, S.; Sundström-Poromaa, I.; Piltonen, T.T.; et al. Elevated prenatal anti-Müllerian hormone reprograms the fetus and induces polycystic ovary syndrome in adulthood. Nat. Med. 2018, 24, 834–846. [Google Scholar] [CrossRef]
- Rudnicka, E.; Kunicki, M.; Calik-Ksepka, A.; Suchta, K.; Duszewska, A.; Smolarczyk, K.; Smolarczyk, R. Anti-Müllerian Hormone in Pathogenesis, Diagnostic and Treatment of PCOS. Int J. Mol. Sci 2021, 22, 2507. [Google Scholar] [CrossRef]
- Palma, G.A.; Argañaraz, M.E.; Barrera, A.D.; Rodler, D.; Mutto, A.Á.; Sinowatz, F. Biology and Biotechnology of Follicle Development. Sci. World J. 2012, 2012, 938138. [Google Scholar] [CrossRef]
- Almeida, F.; Costermans, N.G.J.; Soede, N.M.; Bunschoten, A.; Keijer, J.; Kemp, B.; Teerds, K.J. Presence of anti-Müllerian hormone (AMH) during follicular development in the porcine ovary. PLoS ONE 2018, 13, e0197894. [Google Scholar] [CrossRef] [Green Version]
- Dewailly, D.; Robin, G.; Peigne, M.; Decanter, C.; Pigny, P.; Catteau-Jonard, S. Interactions between androgens, FSH, anti-Müllerian hormone and estradiol during folliculogenesis in the human normal and polycystic ovary. Hum. Reprod. Update 2016, 22, 709–724. [Google Scholar] [CrossRef] [Green Version]
- Dumesic, D.A.; Oberfield, S.E.; Stener-Victorin, E.; Marshall, J.C.; Laven, J.S.; Legro, R.S. Scientific Statement on the Diagnostic Criteria, Epidemiology, Pathophysiology, and Molecular Genetics of Polycystic Ovary Syndrome. Endocr. Rev. 2015, 36, 487–525. [Google Scholar] [CrossRef] [Green Version]
- Ashraf, S.; Nabi, M.; Rasool, S.u.A.; Rashid, F.; Amin, S. Hyperandrogenism in polycystic ovarian syndrome and role of CYP gene variants: A review. Egypt. J. Med. Hum. Genet. 2019, 20, 25. [Google Scholar] [CrossRef] [Green Version]
- Schiffer, L.; Barnard, L.; Baranowski, E.S.; Gilligan, L.C.; Taylor, A.E.; Arlt, W.; Shackleton, C.H.L.; Storbeck, K.H. Human steroid biosynthesis, metabolism and excretion are differentially reflected by serum and urine steroid metabolomes: A comprehensive review. J. Steroid Biochem. Mol. Biol 2019, 194, 105439. [Google Scholar] [CrossRef] [PubMed]
- Dumontet, T.; Martinez, A. Adrenal androgens, adrenarche, and zona reticularis: A human affair? Mol. Cell Endocrinol. 2021, 528, 111239. [Google Scholar] [CrossRef]
- Taylor, A.E.; Ware, M.A.; Breslow, E.; Pyle, L.; Severn, C.; Nadeau, K.J.; Chan, C.L.; Kelsey, M.M.; Cree-Green, M. 11-Oxyandrogens in Adolescents with Polycystic Ovary Syndrome. J. Endocr. Soc. 2022, 6, bvac037. [Google Scholar] [CrossRef]
- O’Reilly, M.W.; Kempegowda, P.; Jenkinson, C.; Taylor, A.E.; Quanson, J.L.; Storbeck, K.H.; Arlt, W. 11-Oxygenated C19 Steroids Are the Predominant Androgens in Polycystic Ovary Syndrome. J. Clin. Endocrinol. Metab. 2017, 102, 840–848. [Google Scholar] [CrossRef]
- Barber, T.M.; Hanson, P.; Weickert, M.O.; Franks, S. Obesity and Polycystic Ovary Syndrome: Implications for Pathogenesis and Novel Management Strategies. Clin. Med. Insights Reprod. Health 2019, 13, 1179558119874042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jamil, A.S.; Alalaf, S.K.; Al-Tawil, N.G.; Al-Shawaf, T. A case–control observational study of insulin resistance and metabolic syndrome among the four phenotypes of polycystic ovary syndrome based on Rotterdam criteria. Reprod. Health 2015, 12, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ding, H.; Zhang, J.; Zhang, F.; Zhang, S.; Chen, X.; Liang, W.; Xie, Q. Resistance to the Insulin and Elevated Level of Androgen: A Major Cause of Polycystic Ovary Syndrome. Front. Endocrinol. 2021, 12, 741764. [Google Scholar] [CrossRef] [PubMed]
- Livadas, S.; Anagnostis, P.; Bosdou, J.K.; Bantouna, D.; Paparodis, R. Polycystic ovary syndrome and type 2 diabetes mellitus: A state-of-the-art review. World J. Diabetes 2022, 13, 5–26. [Google Scholar] [CrossRef]
- Cioana, M.; Deng, J.; Nadarajah, A.; Hou, M.; Qiu, Y.; Chen, S.S.J.; Rivas, A.; Banfield, L.; Alfaraidi, H.; Alotaibi, A.; et al. Prevalence of Polycystic Ovary Syndrome in Patients with Pediatric Type 2 Diabetes: A Systematic Review and Meta-analysis. JAMA Netw. Open 2022, 5, e2147454. [Google Scholar] [CrossRef]
- Krentowska, A.; Łebkowska, A.; Jacewicz-Święcka, M.; Hryniewicka, J.; Leśniewska, M.; Adamska, A.; Kowalska, I. Metabolic syndrome and the risk of cardiovascular complications in young patients with different phenotypes of polycystic ovary syndrome. Endocrine 2021, 72, 400–410. [Google Scholar] [CrossRef]
- Subramanian, A.; Anand, A.; Adderley, N.J.; Okoth, K.; Toulis, K.A.; Gokhale, K.; Sainsbury, C.; O’Reilly, M.W.; Arlt, W.; Nirantharakumar, K. Increased COVID-19 infections in women with polycystic ovary syndrome: A population-based study. Eur. J. Endocrinol. 2021, 184, 637–645. [Google Scholar] [CrossRef]
- de Medeiros, S.F.; Yamamoto, M.M.W.; de Medeiros, M.A.S.; Yamamoto, A.K.L.W.; Barbosa, B.B. Polycystic ovary syndrome and risks for COVID-19 infection: A comprehensive review. Rev. Endocr. Metab. Disord. 2022, 23, 251–264. [Google Scholar] [CrossRef]
- Kho, Z.Y.; Lal, S.K. The Human Gut Microbiome—A Potential Controller of Wellness and Disease. Front. Microbiol. 2018, 9, 1835. [Google Scholar] [CrossRef] [Green Version]
- Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
- Amabebe, E.; Anumba, D.O.C. Female Gut and Genital Tract Microbiota-Induced Crosstalk and Differential Effects of Short-Chain Fatty Acids on Immune Sequelae. Front. Immunol. 2020, 11, 2184. [Google Scholar] [CrossRef]
- Pickard, J.M.; Zeng, M.Y.; Caruso, R.; Núñez, G. Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease. Immunol. Rev. 2017, 279, 70–89. [Google Scholar] [CrossRef]
- Lazar, V.; Ditu, L.-M.; Pircalabioru, G.G.; Gheorghe, I.; Curutiu, C.; Holban, A.M.; Picu, A.; Petcu, L.; Chifiriuc, M.C. Aspects of Gut Microbiota and Immune System Interactions in Infectious Diseases, Immunopathology, and Cancer. Front. Immunol. 2018, 9, 1830. [Google Scholar] [CrossRef] [Green Version]
- Zhao, X.; Jiang, Y.; Xi, H.; Chen, L.; Feng, X. Exploration of the Relationship Between Gut Microbiota and Polycystic Ovary Syndrome (PCOS): A Review. Geburtshilfe Frauenheilkd 2020, 80, 161–171. [Google Scholar] [CrossRef] [Green Version]
- Giampaolino, P.; Foreste, V.; Di Filippo, C.; Gallo, A.; Mercorio, A.; Serafino, P.; Improda, F.P.; Verrazzo, P.; Zara, G.; Buonfantino, C.; et al. Microbiome and PCOS: State-of-Art and Future Aspects. Int. J. Mol. Sci. 2021, 22, 2048. [Google Scholar] [CrossRef]
- Gu, Y.; Zhou, G.; Zhou, F.; Li, Y.; Wu, Q.; He, H.; Zhang, Y.; Ma, C.; Ding, J.; Hua, K. Gut and Vaginal Microbiomes in PCOS: Implications for Women’s Health. Front. Endocrinol. 2022, 13, 808508. [Google Scholar] [CrossRef]
- Graham, M.E.; Herbert, W.G.; Song, S.D.; Raman, H.N.; Zhu, J.E.; Gonzalez, P.E.; Walther-António, M.R.S.; Tetel, M.J. Gut and vaginal microbiomes on steroids: Implications for women’s health. Trends Endocrinol. Metab. 2021, 32, 554–565. [Google Scholar] [CrossRef]
- Thackray, V.G. Sex, Microbes, and Polycystic Ovary Syndrome. Trends Endocrinol. Metab. 2019, 30, 54–65. [Google Scholar] [CrossRef]
- Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [Green Version]
- Chetta, P.; Zadra, G. Metabolic reprogramming as an emerging mechanism of resistance to endocrine therapies in prostate cancer. Cancer Drug Resist. 2021, 4, 143–162. [Google Scholar] [CrossRef]
- Saito, K.; Matsuzaki, T.; Iwasa, T.; Miyado, M.; Saito, H.; Hasegawa, T.; Homma, K.; Inoue, E.; Miyashiro, Y.; Kubota, T.; et al. Steroidogenic pathways involved in androgen biosynthesis in eumenorrheic women and patients with polycystic ovary syndrome. J. Steroid Biochem. Mol. Biol. 2016, 158, 31–37. [Google Scholar] [CrossRef] [PubMed]
- Crommen, S.; Simon, M.C. Microbial Regulation of Glucose Metabolism and Insulin Resistance. Genes 2017, 9, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saad, M.J.; Santos, A.; Prada, P.O. Linking Gut Microbiota and Inflammation to Obesity and Insulin Resistance. Physiology 2016, 31, 283–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pedersen, H.K.; Gudmundsdottir, V.; Nielsen, H.B.; Hyotylainen, T.; Nielsen, T.; Jensen, B.A.; Forslund, K.; Hildebrand, F.; Prifti, E.; Falony, G.; et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 2016, 535, 376–381. [Google Scholar] [CrossRef] [PubMed]
- Sanna, S.; van Zuydam, N.R.; Mahajan, A.; Kurilshikov, A.; Vich Vila, A.; Võsa, U.; Mujagic, Z.; Masclee, A.A.M.; Jonkers, D.; Oosting, M.; et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat. Genet. 2019, 51, 600–605. [Google Scholar] [CrossRef]
- Sun, L.; Xie, C.; Wang, G.; Wu, Y.; Wu, Q.; Wang, X.; Liu, J.; Deng, Y.; Xia, J.; Chen, B.; et al. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat. Med. 2018, 24, 1919–1929. [Google Scholar] [CrossRef]
- Tremellen, K.; Pearce, K. Dysbiosis of Gut Microbiota (DOGMA)--a novel theory for the development of Polycystic Ovarian Syndrome. Med. Hypotheses 2012, 79, 104–112. [Google Scholar] [CrossRef]
- Sun, L.; Hu, W.; Liu, Q.; Hao, Q.; Sun, B.; Zhang, Q.; Mao, S.; Qiao, J.; Yan, X. Metabonomics reveals plasma metabolic changes and inflammatory marker in polycystic ovary syndrome patients. J. Proteome Res. 2012, 11, 2937–2946. [Google Scholar] [CrossRef]
- Zhang, D.; Zhang, L.; Yue, F.; Zheng, Y.; Russell, R. Serum zonulin is elevated in women with polycystic ovary syndrome and correlates with insulin resistance and severity of anovulation. Eur. J. Endocrinol. 2015, 172, 29–36. [Google Scholar] [CrossRef] [Green Version]
- Yurtdas, G.; Akdevelioglu, Y. A New Approach to Polycystic Ovary Syndrome: The Gut Microbiota. J. Am. Coll. Nutr. 2020, 39, 371–382. [Google Scholar] [CrossRef]
- Lim, M.Y.; You, H.J.; Yoon, H.S.; Kwon, B.; Lee, J.Y.; Lee, S.; Song, Y.M.; Lee, K.; Sung, J.; Ko, G. The effect of heritability and host genetics on the gut microbiota and metabolic syndrome. Gut 2017, 66, 1031–1038. [Google Scholar] [CrossRef]
- Lindheim, L.; Bashir, M.; Munzker, J.; Trummer, C.; Zachhuber, V.; Leber, B.; Horvath, A.; Pieber, T.R.; Gorkiewicz, G.; Stadlbauer, V.; et al. Alterations in Gut Microbiome Composition and Barrier Function Are Associated with Reproductive and Metabolic Defects in Women with Polycystic Ovary Syndrome (PCOS): A Pilot Study. PLoS ONE 2017, 12, e0168390. [Google Scholar] [CrossRef] [Green Version]
- Liu, R.; Zhang, C.; Shi, Y.; Zhang, F.; Li, L.; Wang, X.; Ling, Y.; Fu, H.; Dong, W.; Shen, J.; et al. Dysbiosis of Gut Microbiota Associated with Clinical Parameters in Polycystic Ovary Syndrome. Front. Microbiol. 2017, 8, 324. [Google Scholar] [CrossRef]
- Torres, P.J.; Siakowska, M.; Banaszewska, B.; Pawelczyk, L.; Duleba, A.J.; Kelley, S.T.; Thackray, V.G. Gut Microbial Diversity in Women with Polycystic Ovary Syndrome Correlates with Hyperandrogenism. J. Clin. Endocrinol. Metab. 2018, 103, 1502–1511. [Google Scholar] [CrossRef]
- Qi, X.; Yun, C.; Sun, L.; Xia, J.; Wu, Q.; Wang, Y.; Wang, L.; Zhang, Y.; Liang, X.; Wang, L.; et al. Gut microbiota-bile acid-interleukin-22 axis orchestrates polycystic ovary syndrome. Nat. Med. 2019, 25, 1225–1233. [Google Scholar] [CrossRef]
- Ley, R.E. Gut microbiota in 2015: Prevotella in the gut: Choose carefully. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 69–70. [Google Scholar] [CrossRef]
- Zeng, B.; Lai, Z.; Sun, L.; Zhang, Z.; Yang, J.; Li, Z.; Lin, J.; Zhang, Z. Structural and functional profiles of the gut microbial community in polycystic ovary syndrome with insulin resistance (IR-PCOS): A pilot study. Res. Microbiol. 2019, 170, 43–52. [Google Scholar] [CrossRef]
- Li, Y.; Chen, C.; Ma, Y.; Xiao, J.; Luo, G.; Li, Y.; Wu, D. Multi-system reproductive metabolic disorder: Significance for the pathogenesis and therapy of polycystic ovary syndrome (PCOS). Life Sci. 2019, 228, 167–175. [Google Scholar] [CrossRef]
- Lang, U.E.; Beglinger, C.; Schweinfurth, N.; Walter, M.; Borgwardt, S. Nutritional aspects of depression. Cell Physiol. Biochem. 2015, 37, 1029–1043. [Google Scholar] [CrossRef]
- Lin, T.; Li, S.; Xu, H.; Zhou, H.; Feng, R.; Liu, W.; Sun, Y.; Ma, J. Gastrointestinal hormone secretion in women with polycystic ovary syndrome: An observational study. Hum. Reprod. 2015, 30, 2639–2644. [Google Scholar] [CrossRef]
- Falony, G.; Joossens, M.; Vieira-Silva, S.; Wang, J.; Darzi, Y.; Faust, K.; Kurilshikov, A.; Bonder, M.J.; Valles-Colomer, M.; Vandeputte, D.; et al. Population-level analysis of gut microbiome variation. Science 2016, 352, 560–564. [Google Scholar] [CrossRef] [PubMed]
- Haro, C.; Rangel-Zuniga, O.A.; Alcala-Diaz, J.F.; Gomez-Delgado, F.; Perez-Martinez, P.; Delgado-Lista, J.; Quintana-Navarro, G.M.; Landa, B.B.; Navas-Cortes, J.A.; Tena-Sempere, M.; et al. Intestinal Microbiota Is Influenced by Gender and Body Mass Index. PLoS ONE 2016, 11, e0154090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, Y.; Qi, Y.; Yang, X.; Zhao, L.; Wen, S.; Liu, Y.; Tang, L. Association between Polycystic Ovary Syndrome and Gut Microbiota. PLoS ONE 2016, 11, e0153196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, T.; Long, W.; Zhang, C.; Liu, S.; Zhao, L.; Hamaker, B.R. Fiber-utilizing capacity varies in Prevotella- versus Bacteroides-dominated gut microbiota. Sci. Rep. 2017, 7, 2594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dominianni, C.; Sinha, R.; Goedert, J.J.; Pei, Z.; Yang, L.; Hayes, R.B.; Ahn, J. Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PLoS ONE 2015, 10, e0124599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harada, N.; Hanaoka, R.; Horiuchi, H.; Kitakaze, T.; Mitani, T.; Inui, H.; Yamaji, R. Castration influences intestinal microflora and induces abdominal obesity in high-fat diet-fed mice. Sci. Rep. 2016, 6, 23001. [Google Scholar] [CrossRef]
- Barrea, L.; Marzullo, P.; Muscogiuri, G.; Di Somma, C.; Scacchi, M.; Orio, F.; Aimaretti, G.; Colao, A.; Savastano, S. Source and amount of carbohydrate in the diet and inflammation in women with polycystic ovary syndrome. Nutr. Res. Rev. 2018, 31, 291–301. [Google Scholar] [CrossRef]
- Choi, S.; Hwang, Y.J.; Shin, M.J.; Yi, H. Difference in the Gut Microbiome between Ovariectomy-Induced Obesity and Diet-Induced Obesity. J. Microbiol. Biotechnol. 2017, 27, 2228–2236. [Google Scholar] [CrossRef]
- He, F.; Li, Y. The gut microbial composition in polycystic ovary syndrome with insulin resistance: Findings from a normal-weight population. J. Ovarian Res. 2021, 14, 50. [Google Scholar] [CrossRef]
- Qi, X.; Yun, C.; Liao, B.; Qiao, J.; Pang, Y. The therapeutic effect of interleukin-22 in high androgen-induced polycystic ovary syndrome. J. Endocrinol. 2020, 245, 281–289. [Google Scholar] [CrossRef]
- Kriebs, A. IL-22 links gut microbiota to PCOS. Nat. Rev. Endocrinol. 2019, 15, 565. [Google Scholar] [CrossRef]
- Wang, L.; Zhou, J.; Gober, H.J.; Leung, W.T.; Huang, Z.; Pan, X.; Li, C.; Zhang, N.; Wang, L. Alterations in the intestinal microbiome associated with PCOS affect the clinical phenotype. Biomed. Pharm. 2021, 133, 110958. [Google Scholar] [CrossRef]
- Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Backhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Sun, Z.; Jiang, S.; Bai, X.; Ma, C.; Peng, Q.; Chen, K.; Chang, H.; Fang, T.; Zhang, H. Probiotic Bifidobacterium lactis V9 Regulates the Secretion of Sex Hormones in Polycystic Ovary Syndrome Patients through the Gut-Brain Axis. mSystems 2019, 4, e00017-19. [Google Scholar] [CrossRef] [Green Version]
- Chambers, E.S.; Viardot, A.; Psichas, A.; Morrison, D.J.; Murphy, K.G.; Zac-Varghese, S.E.; MacDougall, K.; Preston, T.; Tedford, C.; Finlayson, G.S.; et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 2015, 64, 1744–1754. [Google Scholar] [CrossRef] [Green Version]
- Tolhurst, G.; Heffron, H.; Lam, Y.S.; Parker, H.E.; Habib, A.M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F.M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012, 61, 364–371. [Google Scholar] [CrossRef] [Green Version]
- Kluge, M.; Schussler, P.; Schmidt, D.; Uhr, M.; Steiger, A. Ghrelin suppresses secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) in women. J. Clin. Endocrinol. Metab. 2012, 97, E448–E451. [Google Scholar] [CrossRef] [Green Version]
- Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-kappaB signaling in inflammation. Signal. Transduct Target. 2017, 2, 17023. [Google Scholar] [CrossRef] [Green Version]
- Cardoso, N.S.; Ribeiro, V.B.; Dutra, S.G.V.; Ferriani, R.A.; Gastaldi, A.C.; Araujo, J.E.; Souza, H.C.D. Polycystic ovary syndrome associated with increased adiposity interferes with serum levels of TNF-alpha and IL-6 differently from leptin and adiponectin. Arch. Endocrinol. Metab. 2020, 64, 4–10. [Google Scholar] [CrossRef] [Green Version]
- McCartney, C.R.; Marshall, J.C. Clinical Practice. Polycystic Ovary Syndrome. N. Engl. J. Med. 2016, 375, 54–64. [Google Scholar] [CrossRef]
- Dumesic, D.A.; Akopians, A.L.; Madrigal, V.K.; Ramirez, E.; Margolis, D.J.; Sarma, M.K.; Thomas, A.M.; Grogan, T.R.; Haykal, R.; Schooler, T.A.; et al. Hyperandrogenism Accompanies Increased Intra-Abdominal Fat Storage in Normal Weight Polycystic Ovary Syndrome Women. J. Clin. Endocrinol. Metab. 2016, 101, 4178–4188. [Google Scholar] [CrossRef] [PubMed]
- Martinez Leo, E.E.; Segura Campos, M.R. Effect of ultra-processed diet on gut microbiota and thus its role in neurodegenerative diseases. Nutrition 2020, 71, 110609. [Google Scholar] [CrossRef] [PubMed]
- Rajska, A.; Buszewska-Forajta, M.; Rachon, D.; Markuszewski, M.J. Metabolomic Insight into Polycystic Ovary Syndrome-An Overview. Int. J. Mol. Sci. 2020, 21, 4853. [Google Scholar] [CrossRef] [PubMed]
- Alhilali, M.J.; Parham, A.; Attaranzadeh, A.; Amirian, M.; Azizzadeh, M. Prognostic role of follicular fluid tumor necrosis factor alpha in the risk of early ovarian hyperstimulation syndrome. BMC Pregnancy Childbirth 2020, 20, 691. [Google Scholar] [CrossRef] [PubMed]
- Zhu, S.; Zhang, B.; Jiang, X.; Li, Z.; Zhao, S.; Cui, L.; Chen, Z.J. Metabolic disturbances in non-obese women with polycystic ovary syndrome: A systematic review and meta-analysis. Fertil. Steril. 2019, 111, 168–177. [Google Scholar] [CrossRef] [Green Version]
- Kelley, S.T.; Skarra, D.V.; Rivera, A.J.; Thackray, V.G. The Gut Microbiome Is Altered in a Letrozole-Induced Mouse Model of Polycystic Ovary Syndrome. PLoS ONE 2016, 11, e0146509. [Google Scholar] [CrossRef] [Green Version]
- Chu, W.; Han, Q.; Xu, J.; Wang, J.; Sun, Y.; Li, W.; Chen, Z.J.; Du, Y. Metagenomic analysis identified microbiome alterations and pathological association between intestinal microbiota and polycystic ovary syndrome. Fertil. Steril. 2020, 113, 1286–1298.e1284. [Google Scholar] [CrossRef]
- Chen, F.; Chen, Z.; Chen, M.; Chen, G.; Huang, Q.; Yang, X.; Yin, H.; Chen, L.; Zhang, W.; Lin, H.; et al. Reduced stress-associated FKBP5 DNA methylation together with gut microbiota dysbiosis is linked with the progression of obese PCOS patients. NPJ Biofilms Microbiomes 2021, 7, 60. [Google Scholar] [CrossRef]
- Mammadova, G.; Ozkul, C.; Yilmaz Isikhan, S.; Acikgoz, A.; Yildiz, B.O. Characterization of gut microbiota in polycystic ovary syndrome: Findings from a lean population. Eur. J. Clin. Invest. 2021, 51, e13417. [Google Scholar] [CrossRef]
- Tu, Y.; Zheng, G.; Ding, G.; Wu, Y.; Xi, J.; Ge, Y.; Gu, H.; Wang, Y.; Sheng, J.; Liu, X.; et al. Comparative Analysis of Lower Genital Tract Microbiome Between PCOS and Healthy Women. Front. Physiol 2020, 11, 1108. [Google Scholar] [CrossRef]
- Gupta, S.; Kakkar, V.; Bhushan, I. Crosstalk between Vaginal Microbiome and Female Health: A review. Microb. Pathog. 2019, 136, 103696. [Google Scholar] [CrossRef]
- Al-Memar, M.; Bobdiwala, S.; Fourie, H.; Mannino, R.; Lee, Y.S.; Smith, A.; Marchesi, J.R.; Timmerman, D.; Bourne, T.; Bennett, P.R.; et al. The association between vaginal bacterial composition and miscarriage: A nested case-control study. BJOG 2020, 127, 264–274. [Google Scholar] [CrossRef] [Green Version]
- Peelen, M.J.; Luef, B.M.; Lamont, R.F.; de Milliano, I.; Jensen, J.S.; Limpens, J.; Hajenius, P.J.; Jorgensen, J.S.; Menon, R.; Group, P.B.W. The influence of the vaginal microbiota on preterm birth: A systematic review and recommendations for a minimum dataset for future research. Placenta 2019, 79, 30–39. [Google Scholar] [CrossRef] [Green Version]
- Coudray, M.S.; Madhivanan, P. Bacterial vaginosis-A brief synopsis of the literature. Eur. J. Obs. Gynecol. Reprod. Biol. 2020, 245, 143–148. [Google Scholar] [CrossRef]
- Koedooder, R.; Singer, M.; Schoenmakers, S.; Savelkoul, P.H.M.; Morre, S.A.; de Jonge, J.D.; Poort, L.; Cuypers, W.; Beckers, N.G.M.; Broekmans, F.J.M.; et al. The vaginal microbiome as a predictor for outcome of in vitro fertilization with or without intracytoplasmic sperm injection: A prospective study. Hum. Reprod. 2019, 34, 1042–1054. [Google Scholar] [CrossRef]
- Hong, X.; Qin, P.; Huang, K.; Ding, X.; Ma, J.; Xuan, Y.; Zhu, X.; Peng, D.; Wang, B. Association between polycystic ovary syndrome and the vaginal microbiome: A case-control study. Clin. Endocrinol. 2020, 93, 52–60. [Google Scholar] [CrossRef]
- Sarenac, T.M.; Mikov, M. Bile Acid Synthesis: From Nature to the Chemical Modification and Synthesis and Their Applications as Drugs and Nutrients. Front. Pharm. 2018, 9, 939. [Google Scholar] [CrossRef]
- Wahlstrom, A.; Sayin, S.I.; Marschall, H.U.; Backhed, F. Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef] [Green Version]
- Jia, E.T.; Liu, Z.Y.; Pan, M.; Lu, J.F.; Ge, Q.Y. Regulation of bile acid metabolism-related signaling pathways by gut microbiota in diseases. J. Zhejiang Univ. Sci. B 2019, 20, 781–792. [Google Scholar] [CrossRef]
- Kong, B.; Wang, L.; Chiang, J.Y.; Zhang, Y.; Klaassen, C.D.; Guo, G.L. Mechanism of tissue-specific farnesoid X receptor in suppressing the expression of genes in bile-acid synthesis in mice. Hepatology 2012, 56, 1034–1043. [Google Scholar] [CrossRef]
- Fang, S.; Suh, J.M.; Reilly, S.M.; Yu, E.; Osborn, O.; Lackey, D.; Yoshihara, E.; Perino, A.; Jacinto, S.; Lukasheva, Y.; et al. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat. Med. 2015, 21, 159–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pathak, P.; Xie, C.; Nichols, R.G.; Ferrell, J.M.; Boehme, S.; Krausz, K.W.; Patterson, A.D.; Gonzalez, F.J.; Chiang, J.Y.L. Intestine farnesoid X receptor agonist and the gut microbiota activate G-protein bile acid receptor-1 signaling to improve metabolism. Hepatology 2018, 68, 1574–1588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Trabelsi, M.S.; Daoudi, M.; Prawitt, J.; Ducastel, S.; Touche, V.; Sayin, S.I.; Perino, A.; Brighton, C.A.; Sebti, Y.; Kluza, J.; et al. Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine L cells. Nat. Commun. 2015, 6, 7629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, B.; Shen, S.; Gu, T.; Hong, T.; Liu, J.; Sun, J.; Wang, H.; Bi, Y.; Zhu, D. Increased circulating conjugated primary bile acids are associated with hyperandrogenism in women with polycystic ovary syndrome. J. Steroid Biochem. Mol. Biol. 2019, 189, 171–175. [Google Scholar] [CrossRef]
- He, J.; Zhang, P.; Shen, L.; Niu, L.; Tan, Y.; Chen, L.; Zhao, Y.; Bai, L.; Hao, X.; Li, X.; et al. Short-Chain Fatty Acids and Their Association with Signalling Pathways in Inflammation, Glucose and Lipid Metabolism. Int. J. Mol. Sci. 2020, 21, 6356. [Google Scholar] [CrossRef]
- Murugesan, S.; Nirmalkar, K.; Hoyo-Vadillo, C.; Garcia-Espitia, M.; Ramirez-Sanchez, D.; Garcia-Mena, J. Gut microbiome production of short-chain fatty acids and obesity in children. Eur. J. Clin. Microbiol. Infect. Dis 2018, 37, 621–625. [Google Scholar] [CrossRef]
- Remely, M.; Haslberger, A.G. The microbial epigenome in metabolic syndrome. Mol. Asp. Med. 2017, 54, 71–77. [Google Scholar] [CrossRef]
- Ma, C.; Peng, Q.; Jiang, S.; Zhang, J.; Chen, K.; Fang, Y. Probiotic Bifidobacterium lactis V9 regulates the intestinal microbiome in patients with polycystic ovary syndrome. Chin. Sci. Bull. 2019, 64, 360–368. [Google Scholar] [CrossRef] [Green Version]
- Li, S.; Chu, Q.; Ma, J.; Sun, Y.; Tao, T.; Huang, R.; Liao, Y.; Yue, J.; Zheng, J.; Wang, L.; et al. Discovery of Novel Lipid Profiles in PCOS: Do Insulin and Androgen Oppositely Regulate Bioactive Lipid Production? J. Clin. Endocrinol. Metab. 2017, 102, 810–821. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Xie, L.M.; Song, J.L.; Yau, L.F.; Mi, J.N.; Zhang, C.R.; Wu, W.T.; Lai, M.H.; Jiang, Z.H.; Wang, J.R.; et al. Alterations of Sphingolipid Metabolism in Different Types of Polycystic Ovary Syndrome. Sci. Rep. 2019, 9, 3204. [Google Scholar] [CrossRef]
- Jove, M.; Pradas, I.; Naudi, A.; Rovira-Llopis, S.; Banuls, C.; Rocha, M.; Portero-Otin, M.; Hernandez-Mijares, A.; Victor, V.M.; Pamplona, R. Lipidomics reveals altered biosynthetic pathways of glycerophospholipids and cell signaling as biomarkers of the polycystic ovary syndrome. Oncotarget 2018, 9, 4522–4536. [Google Scholar] [CrossRef] [Green Version]
- Johnson, E.L.; Heaver, S.L.; Waters, J.L.; Kim, B.I.; Bretin, A.; Goodman, A.L.; Gewirtz, A.T.; Worgall, T.S.; Ley, R.E. Sphingolipids produced by gut bacteria enter host metabolic pathways impacting ceramide levels. Nat. Commun. 2020, 11, 2471. [Google Scholar] [CrossRef]
- Kayser, B.D.; Prifti, E.; Lhomme, M.; Belda, E.; Dao, M.C.; Aron-Wisnewsky, J.; Consortium, M.I.-O.; Kontush, A.; Zucker, J.D.; Rizkalla, S.W.; et al. Elevated serum ceramides are linked with obesity-associated gut dysbiosis and impaired glucose metabolism. Metabolomics 2019, 15, 140. [Google Scholar] [CrossRef]
- Schiattarella, G.G.; Sannino, A.; Toscano, E.; Giugliano, G.; Gargiulo, G.; Franzone, A.; Trimarco, B.; Esposito, G.; Perrino, C. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: A systematic review and dose-response meta-analysis. Eur. Heart J. 2017, 38, 2948–2956. [Google Scholar] [CrossRef] [Green Version]
- Tang, W.H.W.; Li, D.Y.; Hazen, S.L. Dietary metabolism, the gut microbiome, and heart failure. Nat. Rev. Cardiol 2019, 16, 137–154. [Google Scholar] [CrossRef]
- Lemaitre, R.N.; Jensen, P.N.; Wang, Z.; Fretts, A.M.; McKnight, B.; Nemet, I.; Biggs, M.L.; Sotoodehnia, N.; de Oliveira Otto, M.C.; Psaty, B.M.; et al. Association of Trimethylamine N-Oxide and Related Metabolites in Plasma and Incident Type 2 Diabetes: The Cardiovascular Health Study. JAMA Netw. Open 2021, 4, e2122844. [Google Scholar] [CrossRef]
- Shan, Z.; Sun, T.; Huang, H.; Chen, S.; Chen, L.; Luo, C.; Yang, W.; Yang, X.; Yao, P.; Cheng, J.; et al. Association between microbiota-dependent metabolite trimethylamine-N-oxide and type 2 diabetes. Am. J. Clin. Nutr. 2017, 106, 888–894. [Google Scholar] [CrossRef] [Green Version]
- Heianza, Y.; Sun, D.; Li, X.; DiDonato, J.A.; Bray, G.A.; Sacks, F.M.; Qi, L. Gut microbiota metabolites, amino acid metabolites and improvements in insulin sensitivity and glucose metabolism: The POUNDS Lost trial. Gut 2019, 68, 263–270. [Google Scholar] [CrossRef]
- Chen, S.; Henderson, A.; Petriello, M.C.; Romano, K.A.; Gearing, M.; Miao, J.; Schell, M.; Sandoval-Espinola, W.J.; Tao, J.; Sha, B.; et al. Trimethylamine N-Oxide Binds and Activates PERK to Promote Metabolic Dysfunction. Cell Metab. 2019, 30, 1141–1151.e1145. [Google Scholar] [CrossRef]
- Eyupoglu, N.D.; Caliskan Guzelce, E.; Acikgoz, A.; Uyanik, E.; Bjorndal, B.; Berge, R.K.; Svardal, A.; Yildiz, B.O. Circulating gut microbiota metabolite trimethylamine N-oxide and oral contraceptive use in polycystic ovary syndrome. Clin. Endocrinol. 2019, 91, 810–815. [Google Scholar] [CrossRef]
- Annunziata, G.; Ciampaglia, R.; Capo, X.; Guerra, F.; Sureda, A.; Tenore, G.C.; Novellino, E. Polycystic ovary syndrome and cardiovascular risk. Could trimethylamine N-oxide (TMAO) be a major player? A potential upgrade forward in the DOGMA theory. Biomed. Pharm. 2021, 143, 112171. [Google Scholar] [CrossRef] [PubMed]
- Lagana, A.S.; Rossetti, P.; Buscema, M.; La Vignera, S.; Condorelli, R.A.; Gullo, G.; Granese, R.; Triolo, O. Metabolism and Ovarian Function in PCOS Women: A Therapeutic Approach with Inositols. Int. J. Endocrinol. 2016, 2016, 6306410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abraham Gnanadass, S.; Divakar Prabhu, Y.; Valsala Gopalakrishnan, A. Association of metabolic and inflammatory markers with polycystic ovarian syndrome (PCOS): An update. Arch. Gynecol. Obs. 2021, 303, 631–643. [Google Scholar] [CrossRef] [PubMed]
- Cassar, S.; Misso, M.L.; Hopkins, W.G.; Shaw, C.S.; Teede, H.J.; Stepto, N.K. Insulin resistance in polycystic ovary syndrome: A systematic review and meta-analysis of euglycaemic-hyperinsulinaemic clamp studies. Hum. Reprod. 2016, 31, 2619–2631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barber, T.M.; Dimitriadis, G.K.; Andreou, A.; Franks, S. Polycystic ovary syndrome: Insight into pathogenesis and a common association with insulin resistance. Clin. Med. 2016, 16, 262–266. [Google Scholar] [CrossRef] [Green Version]
- Yang, P.K.; Chou, C.H.; Huang, C.C.; Wen, W.F.; Chen, H.F.; Shun, C.T.; Ho, H.N.; Chen, M.J. Obesity alters ovarian folliculogenesis through disrupted angiogenesis from increased IL-10 production. Mol. Metab. 2021, 49, 101189. [Google Scholar] [CrossRef]
- Lerner, A.; Kewada, D.; Ahmed, A.; Hardy, K.; Christian, M.; Franks, S. Androgen Reduces Mitochondrial Respiration in Mouse Brown Adipocytes: A Model for Disordered Energy Balance in Polycystic Ovary Syndrome. Int. J. Mol. Sci. 2020, 22, 243. [Google Scholar] [CrossRef]
- Zhou, J.; Huang, X.; Xue, B.; Wei, Y.; Hua, F. Bioinformatics analysis of the molecular mechanism of obesity in polycystic ovary syndrome. Aging 2021, 13, 12631–12640. [Google Scholar] [CrossRef]
- Kany, S.; Vollrath, J.T.; Relja, B. Cytokines in Inflammatory Disease. Int. J. Mol. Sci. 2019, 20, 6008. [Google Scholar] [CrossRef] [Green Version]
- Niu, Z.; Ye, Y.; Xia, L.; Feng, Y.; Zhang, A. Follicular fluid cytokine composition and oocyte quality of polycystic ovary syndrome patients with metabolic syndrome undergoing in vitro fertilization. Cytokine 2017, 91, 180–186. [Google Scholar] [CrossRef]
- Prabhu, Y.D.; Borthakur, A.; Subeka, G.S.; Vellingiri, B.; Valsala Gopalakrishnan, A. Increased pro-inflammatory cytokines in ovary and effect of gamma-linolenic acid on adipose tissue inflammation in a polycystic ovary syndrome model. J. Reprod. Immunol. 2021, 146, 103345. [Google Scholar] [CrossRef]
- Regidor, P.A.; Mueller, A.; Sailer, M.; Gonzalez Santos, F.; Rizo, J.M.; Egea, F.M. Chronic Inflammation in PCOS: The Potential Benefits of Specialized Pro-Resolving Lipid Mediators (SPMs) in the Improvement of the Resolutive Response. Int. J. Mol. Sci. 2020, 22, 384. [Google Scholar] [CrossRef]
- Svendsen, P.F.; Christiansen, M.; Hedley, P.L.; Nilas, L.; Pedersen, S.B.; Madsbad, S. Adipose expression of adipocytokines in women with polycystic ovary syndrome. Fertil. Steril. 2012, 98, 235–241. [Google Scholar] [CrossRef]
- Orostica, L.; Astorga, I.; Plaza-Parrochia, F.; Vera, C.; Garcia, V.; Carvajal, R.; Gabler, F.; Romero, C.; Vega, M. Proinflammatory environment and role of TNF-alpha in endometrial function of obese women having polycystic ovarian syndrome. Int. J. Obes. 2016, 40, 1715–1722. [Google Scholar] [CrossRef]
- Altonen, B.L.; Arreglado, T.M.; Leroux, O.; Murray-Ramcharan, M.; Engdahl, R. Characteristics, comorbidities and survival analysis of young adults hospitalized with COVID-19 in New York City. PLoS ONE 2020, 15, e0243343. [Google Scholar] [CrossRef]
- Ghowsi, M.; Khazali, H.; Sisakhtnezhad, S. Evaluation of TNF-alpha and IL-6 mRNAs expressions in visceral and subcutaneous adipose tissues of polycystic ovarian rats and effects of resveratrol. Iran. J. Basic Med. Sci. 2018, 21, 165–174. [Google Scholar] [CrossRef]
- Adams, J.; Liu, Z.; Ren, Y.A.; Wun, W.S.; Zhou, W.; Kenigsberg, S.; Librach, C.; Valdes, C.; Gibbons, W.; Richards, J. Enhanced Inflammatory Transcriptome in the Granulosa Cells of Women with Polycystic Ovarian Syndrome. J. Clin. Endocrinol. Metab. 2016, 101, 3459–3468. [Google Scholar] [CrossRef] [Green Version]
- Artimani, T.; Karimi, J.; Mehdizadeh, M.; Yavangi, M.; Khanlarzadeh, E.; Ghorbani, M.; Asadi, S.; Kheiripour, N. Evaluation of pro-oxidant-antioxidant balance (PAB) and its association with inflammatory cytokines in polycystic ovary syndrome (PCOS). Gynecol. Endocrinol. 2018, 34, 148–152. [Google Scholar] [CrossRef]
- Zhang, H.; Wang, X.; Xu, J.; Zhu, Y.; Chen, X.; Hu, Y. IL-18 and IL-18 binding protein concentration in ovarian follicular fluid of women with unexplained infertility to PCOS during in vitro fertilization. J. Reprod. Immunol. 2020, 138, 103083. [Google Scholar] [CrossRef]
- Bannigida, D.M.; Nayak, B.S.; Vijayaraghavan, R. Insulin resistance and oxidative marker in women with PCOS. Arch. Physiol. Biochem. 2020, 126, 183–186. [Google Scholar] [CrossRef]
- Su, H.; Lei, C.-T.; Zhang, C. Interleukin-6 Signaling Pathway and Its Role in Kidney Disease: An Update. Front. Immunol. 2017, 8, 405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Graham, A.M.; Rasmussen, J.M.; Rudolph, M.D.; Heim, C.M.; Gilmore, J.H.; Styner, M.; Potkin, S.G.; Entringer, S.; Wadhwa, P.D.; Fair, D.A.; et al. Maternal Systemic Interleukin-6 During Pregnancy Is Associated with Newborn Amygdala Phenotypes and Subsequent Behavior at 2 Years of Age. Biol. Psychiatry 2018, 83, 109–119. [Google Scholar] [CrossRef] [PubMed]
- 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, e0148531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, C.-H.; Moon, J.-W.; Moon, S.Y. The effect of interleukin 6 on controlled ovarian stimulation results and IVF outcome in infertile women with adenomyosis undergoing IVF. Fertil. Steril. 2019, 112, e187. [Google Scholar] [CrossRef]
- Srivastava, S.K.; Bhardwaj, A.; Arora, S.; Tyagi, N.; Singh, A.P.; Carter, J.E.; Scammell, J.G.; Fodstad, O.; Singh, S. Interleukin-8 is a key mediator of FKBP51-induced melanoma growth, angiogenesis and metastasis. Br. J. Cancer 2015, 112, 1772–1781. [Google Scholar] [CrossRef] [Green Version]
- Ali, D.E.; Shah, M.; Ali, A.; Malik, M.O.; Rehman, F.; Badshah, H.; Ehtesham, E.; Vitale, S.G. Treatment with Metformin and Combination of Metformin Plus Pioglitazone on Serum Levels of IL-6 and IL-8 in Polycystic Ovary Syndrome: A Randomized Clinical Trial. Horm. Metab. Res. 2019, 51, 714–722. [Google Scholar] [CrossRef]
- Esposito, K.; Pontillo, A.; Giugliano, F.; Giugliano, G.; Marfella, R.; Nicoletti, G.; Giugliano, D. Association of low interleukin-10 levels with the metabolic syndrome in obese women. J. Clin. Endocrinol. Metab. 2003, 88, 1055–1058. [Google Scholar] [CrossRef]
- Talaat, R.M.; Mohamed, Y.A.; Mohamad, E.H.; Elsharkawy, M.; Guirgis, A.A. Interleukin 10 (-1082 G/A) and (-819 C/T) gene polymorphisms in Egyptian women with polycystic ovary syndrome (PCOS). Meta Gene 2016, 9, 254–258. [Google Scholar] [CrossRef] [Green Version]
- Sylus, A.M.; Nandeesha, H.; Sridhar, M.G.; Chitra, T.; Sreenivasulu, K. Clomiphene citrate increases nitric oxide, interleukin-10 and reduces matrix metalloproteinase-9 in women with polycystic ovary syndrome. Eur. J. Obs. Gynecol. Reprod. Biol. 2018, 228, 27–31. [Google Scholar] [CrossRef]
- Long, X.; Li, R.; Yang, Y.; Qiao, J. Overexpression of IL-18 in the Proliferative Phase Endometrium of Patients with Polycystic Ovary Syndrome. Reprod. Sci. 2017, 24, 252–257. [Google Scholar] [CrossRef]
- Dawood, A.; Alkafrawy, N.; Saleh, S.; Noreldin, R.; Zewain, S. The relationship between IL-18 and atherosclerotic cardiovascular risk in Egyptian lean women with polycystic ovary syndrome. Gynecol. Endocrinol. 2018, 34, 294–297. [Google Scholar] [CrossRef]
- Sathyapalan, T.; Atkin, S.L. Mediators of inflammation in polycystic ovary syndrome in relation to adiposity. Mediat. Inflamm. 2010, 2010, 758656. [Google Scholar] [CrossRef] [Green Version]
- Kalyan, S.; Goshtesabi, A.; Sarray, S.; Joannou, A.; Almawi, W.Y. Assessing C reactive protein/albumin ratio as a new biomarker for polycystic ovary syndrome: A case-control study of women from Bahraini medical clinics. BMJ Open 2018, 8, e021860. [Google Scholar] [CrossRef]
- Elci, E.; Kaya, C.; Cim, N.; Yildizhan, R.; Elci, G.G. Evaluation of cardiac risk marker levels in obese and non-obese patients with polycystic ovaries. Gynecol.Endocrinol. 2017, 33, 43–47. [Google Scholar] [CrossRef]
- Escobar-Morreale, H.F.; Luque-Ramírez, M.; González, F. Circulating inflammatory markers in polycystic ovary syndrome: A systematic review and metaanalysis. Fertil. Steril. 2011, 95, 1048–1058.e1042. [Google Scholar] [CrossRef] [Green Version]
- Fathi, F.H. C-Reactive Protein and Adiposity in Women with Polycystic Ovary Syndrome. Tikrit J. Pure Sci. 2018, 23, 19–27. [Google Scholar]
- Neelaveni, K.; Menon, R.; Sahay, R.K.; Reddy, G.C.. CRP levels and endothelial function in young women with PCOS. J. Evol. Med. Dent. Sci. 2016, 5, 5783–5787. [Google Scholar] [CrossRef]
- Duleba, A.J.; Dokras, A. Is PCOS an inflammatory process? Fertil. Steril. 2012, 97, 7–12. [Google Scholar] [CrossRef] [Green Version]
- Kelly, C.C.; Lyall, H.; Petrie, J.R.; Gould, G.W.; Connell, J.M.; Sattar, N. Low grade chronic inflammation in women with polycystic ovarian syndrome. J. Clin. Endocrinol. Metab. 2001, 86, 2453–2455. [Google Scholar] [CrossRef]
- Engin-Üstün, Y.; Üstün, Y.; Meydanli, M.M.; Kafkasli, A.; Yetkin, G.J.G.e. Are polycystic ovaries associated with cardiovascular disease risk as polycystic ovary syndrome? Gynecol. Endocrinol. 2006, 22, 324–328. [Google Scholar] [CrossRef]
- Cakal, E.; Ustun, Y.; Engin-Ustun, Y.; Ozkaya, M.; Kilinç, M. Serum vaspin and C-reactive protein levels in women with polycystic ovaries and polycystic ovary syndrome. Gynecol. Endocrinol. 2011, 27, 491–495. [Google Scholar] [CrossRef] [PubMed]
- Festa, A.; D’Agostino Jr, R.; Howard, G.; Mykkanen, L.; Tracy, R.P.; Haffner, S.M. Chronic subclinical inflammation as part of the insulin resistance syndrome: The Insulin Resistance Atherosclerosis Study (IRAS). Circulation 2000, 102, 42–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Engeli, S.; Feldpausch, M.; Gorzelniak, K.; Hartwig, F.; Heintze, U.; Janke, J.; Mohlig, M.; Pfeiffer, A.F.; Luft, F.C.; Sharma, A.M. Association between adiponectin and mediators of inflammation in obese women. Diabetes 2003, 52, 942–947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tola, E.N.; Yalcin, S.E.; Dugan, N. The predictive effect of inflammatory markers and lipid accumulation product index on clinical symptoms associated with polycystic ovary syndrome in nonobese adolescents and younger aged women. Eur. J. Obstet. Gynecol. Reprod. Biol. Endocrinol. 2017, 214, 168–172. [Google Scholar] [CrossRef] [PubMed]
- Souza dos Santos, A.C.; Soares, N.P.; Costa, E.C.; de Sá, J.C.F.; Azevedo, G.D.; Lemos, T.M.A.M. The impact of body mass on inflammatory markers and insulin resistance in polycystic ovary syndrome. Gynecol. Endocrinol. 2015, 31, 225–228. [Google Scholar] [CrossRef] [PubMed]
- Orio, F., Jr.; Palomba, S.; Cascella, T.; Di Biase, S.; Manguso, F.; Tauchmanovà, L.; Nardo, L.G.; Labella, D.; Savastano, S.; Russo, T. The increase of leukocytes as a new putative marker of low-grade chronic inflammation and early cardiovascular risk in polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 2005, 90, 2–5. [Google Scholar] [CrossRef] [Green Version]
- Rudnicka, E.; Kunicki, M.; Suchta, K.; Machura, P.; Grymowicz, M.; Smolarczyk, R. Inflammatory markers in women with polycystic ovary syndrome. BioMed Res. Int. 2020, 2020, 4092470. [Google Scholar] [CrossRef] [Green Version]
- Smith, S.B.; Ravel, J. The vaginal microbiota, host defence and reproductive physiology. J. Physiol. 2017, 595, 451–463. [Google Scholar] [CrossRef] [Green Version]
- Dabke, K.; Hendrick, G.; Devkota, S. The gut microbiome and metabolic syndrome. J. Clin. Investig. 2019, 129, 4050–4057. [Google Scholar] [CrossRef]
- Greenbaum, S.; Greenbaum, G.; Moran-Gilad, J.; Weintraub, A.Y. Gynecology. Ecological dynamics of the vaginal microbiome in relation to health and disease. Am. J. Obstet. Gynecol. 2019, 220, 324–335. [Google Scholar] [CrossRef]
- Lu, C.; Wang, H.; Yang, J.; Zhang, X.; Chen, Y.; Feng, R.; Qian, Y. Changes in Vaginal Microbiome Diversity in Women With Polycystic Ovary Syndrome. Front. Cell. Infect. Microbiol. 2021, 11, 755741. [Google Scholar] [CrossRef]
- Xu, J.; Bian, G.; Zheng, M.; Lu, G.; Chan, W.Y.; Li, W.; Yang, K.; Chen, Z.J.; Du, Y.J. Fertility factors affect the vaginal microbiome in women of reproductive age. Am. J. Reprod. Immunol. 2020, 83, e13220. [Google Scholar] [CrossRef] [Green Version]
- Yan, S.-K.; Liu, R.-H.; Jin, H.-Z.; Liu, X.-R.; Ye, J.; Shan, L.; Zhang, W.-D. “Omics” in pharmaceutical research: Overview, applications, challenges, and future perspectives. Chin. J. Nat. Med. 2015, 13, 3–21. [Google Scholar] [CrossRef]
- Ma, X.; Fan, L.; Meng, Y.; Hou, Z.; Mao, Y.-D.; Wang, W.; Ding, W.; Liu, J.-Y.J. Proteomic analysis of human ovaries from normal and polycystic ovarian syndrome. MHR Basic Sci. Reprod. Med. 2007, 13, 527–535. [Google Scholar] [CrossRef]
- Jungert, A.; Frank, J. Intra-Individual Variation and Reliability of Biomarkers of the Antioxidant Defense System by Considering Dietary and Lifestyle Factors in Premenopausal Women. Antioxidants 2021, 10, 448. [Google Scholar] [CrossRef]
- Johnson, C.H.; Gonzalez, F.J. Challenges and opportunities of metabolomics. J. Cell. Physiol. 2012, 227, 2975–2981. [Google Scholar] [CrossRef]
- Azziz, R.; Woods, K.S.; Reyna, R.; Key, T.J.; Knochenhauer, E.S.; Yildiz, B.O. Metabolism. The prevalence and features of the polycystic ovary syndrome in an unselected population. J. Clin. Endocrinol. Metab. 2004, 89, 2745–2749. [Google Scholar] [CrossRef] [Green Version]
- Apridonidze, T.; Essah, P.A.; Iuorno, M.J.; Nestler, J.E.J. Prevalence and characteristics of the metabolic syndrome in women with polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 2005, 90, 1929–1935. [Google Scholar] [CrossRef] [Green Version]
- Carmina, E.; Napoli, N.; Longo, R.; Rini, G.; Lobo, R.J. Metabolic syndrome in polycystic ovary syndrome (PCOS): Lower prevalence in southern Italy than in the USA and the influence of criteria for the diagnosis of PCOS. Eur. J. Endocrinol. 2006, 154, 141–145. [Google Scholar] [CrossRef] [Green Version]
- Ehrmann, D.A.; Liljenquist, D.R.; Kasza, K.; Azziz, R.; Legro, R.S.; Ghazzi, M.N. Prevalence and predictors of the metabolic syndrome in women with polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 2006, 91, 48–53. [Google Scholar] [CrossRef] [Green Version]
- Kekwick, R.A. The electrophoretic analysis of normal human serum. Biochem. J. 1939, 33, 1122. [Google Scholar] [CrossRef] [PubMed]
- Psychogios, N.; Hau, D.D.; Peng, J.; Guo, A.C.; Mandal, R.; Bouatra, S.; Sinelnikov, I.; Krishnamurthy, R.; Eisner, R.; Gautam, B.; et al. The human serum metabolome. PLoS ONE 2011, 6, e16957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, K.; Fang, J.; Jin, J.; Zhu, S.; Xu, X.; Xu, Y.; Ye, B.; Lin, S.-H.; Xu, X.J. Serum metabolomics reveals personalized metabolic patterns for macular neovascular disease patient stratification. J. Proteome Res. 2019, 19, 699–707. [Google Scholar] [CrossRef]
- Jin-Wei, H.; Sa, Y.-H.; Song, J.; Sun, Z. Pseudo-targeted metabolomic analysis of Chinese Herbal Medicines Cangfu Daotan Decoction to improve in vitro fertilization outcomes of patients with Polycystic Ovary Syndrome. Res. Sq. 2022, preprint. [Google Scholar]
- Roberts, L.D.; Souza, A.L.; Gerszten, R.E.; Clish, C.B. Targeted metabolomics. Curr. Protoc. Mol. Biol. 2012, 98, 30. [Google Scholar] [CrossRef]
- Ożegowska, K.; Plewa, S.; Mantaj, U.; Pawelczyk, L.; Matysiak, J. Serum Metabolomics in PCOS Women with Different Body Mass Index. J. Clin. Med. 2021, 10, 2811. [Google Scholar] [CrossRef]
- Buszewska-Forajta, M.; Rachoń, D.; Stefaniak, A.; Wawrzyniak, R.; Konieczna, A.; Kowalewska, A.; Markuszewski, M.J. Identification of the metabolic fingerprints in women with polycystic ovary syndrome using the multiplatform metabolomics technique. J. Steroid Biochem. Mol. Biol. 2019, 186, 176–184. [Google Scholar] [CrossRef]
- Yu, Y.; Tan, P.; Zhuang, Z.; Wang, Z.; Zhu, L.; Qiu, R.; Xu, H. Untargeted metabolomic approach to study the serum metabolites in women with polycystic ovary syndrome. BMC Med. Genom. 2021, 14, 206. [Google Scholar] [CrossRef]
- Liu, L.; Yin, T.-l.; Chen, Y.; Li, Y.; Yin, L.; Ding, J.; Yang, J.; Feng, H.-L. Follicular dynamics of glycerophospholipid and sphingolipid metabolisms in polycystic ovary syndrome patients. J. Steroid Biochem. Mol. Biol. 2019, 185, 142–149. [Google Scholar] [CrossRef]
- Lim, S.; Kakoly, N.; Tan, J.; Fitzgerald, G.; Bahri Khomami, M.; Joham, A.; Cooray, S.; Misso, M.; Norman, R.; Harrison, C. Metabolic syndrome in polycystic ovary syndrome: A systematic review, meta-analysis and meta-regression. Obes. Rev. 2019, 20, 339–352. [Google Scholar] [CrossRef]
- Rosenfield, R.L. The diagnosis of polycystic ovary syndrome in adolescents. Pediatrics 2015, 136, 1154–1165. [Google Scholar] [CrossRef] [Green Version]
- Dunaif, A.; Segal, K.R.; Futterweit, W.; Dobrjansky, A. Profound peripheral insulin resistance, independent of obesity, in polycystic ovary syndrome. Diabetes 1989, 38, 1165–1174. [Google Scholar] [CrossRef]
- Stepto, N.K.; Cassar, S.; Joham, A.E.; Hutchison, S.K.; Harrison, C.L.; Goldstein, R.F.; Teede, H.J. Women with polycystic ovary syndrome have intrinsic insulin resistance on euglycaemic–hyperinsulaemic clamp. Hum. Reprod. 2013, 28, 777–784. [Google Scholar] [CrossRef]
- Willis, D.S.; Watson, H.; Mason, H.D.; Galea, R.; Brincat, M.; Franks, S. Premature response to luteinizing hormone of granulosa cells from anovulatory women with polycystic ovary syndrome: Relevance to mechanism of anovulation. J. Clin. Endocrinol. Metab. 1998, 83, 3984–3991. [Google Scholar] [CrossRef]
- Garg, D.; Tal, R. Inositol treatment and ART outcomes in women with PCOS. Int. J. Endocrinol. 2016, 2016, 1979654. [Google Scholar] [CrossRef] [Green Version]
- Shen, Q.; Xiao, X.; Aierken, A.; Yue, W.; Wu, X.; Liao, M.; Hua, J. The ACE2 expression in Sertoli cells and germ cells may cause male reproductive disorder after SARS-CoV-2 infection. J. Cell. Mol. Med. 2020, 24, 9472–9477. [Google Scholar] [CrossRef]
- Vrbikova, J.; Hill, M.; Bendlova, B.; Grimmichova, T.; Dvorakova, K.; Vondra, K.; Pacini, G. Incretin levels in polycystic ovary syndrome. Eur. J. Endocrinol. 2008, 159, 121–128. [Google Scholar] [CrossRef] [Green Version]
- Essah, P.A.; Wickham, E.P.; Nestler, J.E. The metabolic syndrome in polycystic ovary syndrome. Clin. Obstet. Gynecol. 2007, 50, 205–225. [Google Scholar] [CrossRef]
- Anagnostis, P.; Tarlatzis, B.C.; Kauffman, R.P. Polycystic ovarian syndrome (PCOS): Long-term metabolic consequences. Metabolism 2018, 86, 33–43. [Google Scholar] [CrossRef]
- Deba, Z.; Jambale, T.A.; Swamy, P.G.; Murthy, D.J. Study of levels of malondialdehyde, super oxide dismutase and hs-CRP in serum of non-obese patients with polycystic ovarian syndrome. Int. J. Clin. Biochem. 2017, 4, 191–194. [Google Scholar] [CrossRef]
- Adamska, A.; Łebkowska, A.; Krentowska, A.; Adamski, M.; Kowalska, I. The association between serum ferritin concentration and visceral adiposity estimated by whole-body DXA scan in women with polycystic ovary syndrome. Front. Endocrinol. 2020, 10, 873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Macut, D.; Tziomalos, K.; Božić-Antić, I.; Bjekić-Macut, J.; Katsikis, I.; Papadakis, E.; Andrić, Z.; Panidis, D. Non-alcoholic fatty liver disease is associated with insulin resistance and lipid accumulation product in women with polycystic ovary syndrome. Hum. Reprod. 2016, 31, 1347–1353. [Google Scholar] [CrossRef] [PubMed]
- Vassilatou, E.; Lafoyianni, S.; Vryonidou, A.; Ioannidis, D.; Kosma, L.; Katsoulis, K.; Papavassiliou, E.; Tzavara, I. Increased androgen bioavailability is associated with non-alcoholic fatty liver disease in women with polycystic ovary syndrome. Hum. Reprod. 2010, 25, 212–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, H.; Sprung, V.S.; Pugh, C.J.; Daousi, C.; Irwin, A.; Aziz, N.; Adams, V.L.; Thomas, E.L.; Bell, J.D.; Kemp, G.J. Polycystic ovary syndrome with hyperandrogenism is characterized by an increased risk of hepatic steatosis compared to nonhyperandrogenic PCOS phenotypes and healthy controls, independent of obesity and insulin resistance. J. Clin. Endocrinol. Metab. 2012, 97, 3709–3716. [Google Scholar] [CrossRef] [PubMed]
- Hossain, N.; Stepanova, M.; Afendy, A.; Nader, F.; Younossi, Y.; Rafiq, N.; Goodman, Z.; Younossi, Z.M. Non-alcoholic steatohepatitis (NASH) in patients with polycystic ovarian syndrome (PCOS). Scand. J. Gastroenterol. 2011, 46, 479–484. [Google Scholar] [CrossRef] [PubMed]
- Brzozowska, M.M.; Ostapowicz, G.; Weltman, M.D. An association between non-alcoholic fatty liver disease and polycystic ovarian syndrome. J. Gastroenterol. Hepatol. 2009, 24, 243–247. [Google Scholar] [CrossRef] [PubMed]
- Karoli, R.; Fatima, J.; Chandra, A.; Gupta, U.; Islam, F.-u.; Singh, G. Prevalence of hepatic steatosis in women with polycystic ovary syndrome. J. Hum. Reprod. Sci. 2013, 6, 9. [Google Scholar] [CrossRef]
- Macut, D.; Božić-Antić, I.; Bjekić-Macut, J.; Tziomalos, K. Management of endocrine disease: Polycystic ovary syndrome and nonalcoholic fatty liver disease. Eur. J. Endocrinol. 2017, 177, R145–R158. [Google Scholar] [CrossRef] [Green Version]
- Younossi, Z.M.; Stepanova, M.; Afendy, M.; Fang, Y.; Younossi, Y.; Mir, H.; Srishord, M. Changes in the prevalence of the most common causes of chronic liver diseases in the United States from 1988 to 2008. Clin. Gastroenterol. Hepatol. 2011, 9, 524–530.e521. [Google Scholar] [CrossRef]
- Diamanti-Kandarakis, E.; Dunaif, A. Insulin resistance and the polycystic ovary syndrome revisited: An update on mechanisms and implications. Endocr. Rev. 2012, 33, 981–1030. [Google Scholar] [CrossRef]
- Kim, J.; Kim, D.; Yim, J.; Kang, J.; Han, K.; Kim, S.; Hwang, K.; Ku, S.; Suh, C.; Kim, S. Polycystic ovary syndrome with hyperandrogenism as a risk factor for non-obese non-alcoholic fatty liver disease. Aliment. Pharmacol. Ther. 2017, 45, 1403–1412. [Google Scholar] [CrossRef] [Green Version]
- Wu, J.; Yao, X.-Y.; Shi, R.-X.; Liu, S.-F.; Wang, X.-Y. A potential link between polycystic ovary syndrome and non-alcoholic fatty liver disease: An update meta-analysis. Reprod. Health 2018, 15, 77. [Google Scholar] [CrossRef]
- Zhang, J.; Hu, J.; Zhang, C.; Jiao, Y.; Kong, X.; Wang, W. Analyses of risk factors for polycystic ovary syndrome complicated with non-alcoholic fatty liver disease. Exp. Ther. Med. 2018, 15, 4259–4264. [Google Scholar] [CrossRef] [Green Version]
- Després, J.-P.; Lemieux, I. Abdominal obesity and metabolic syndrome. Nature 2006, 444, 881–887. [Google Scholar] [CrossRef]
- Dawson, A.J.; Sathyapalan, T.; Smithson, J.A.; Vince, R.V.; Coady, A.M.; Ajjan, R.; Kilpatrick, E.S.; Atkin, S.L. A comparison of cardiovascular risk indices in patients with polycystic ovary syndrome with and without coexisting nonalcoholic fatty liver disease. Clin. Endocrinol. 2014, 80, 843–849. [Google Scholar] [CrossRef]
- Gambarin–Gelwan, M.; Kinkhabwala, S.V.; Schiano, T.D.; Bodian, C.; Yeh, H.C.; Futterweit, W. Prevalence of nonalcoholic fatty liver disease in women with polycystic ovary syndrome. Clin. Gastroenterol. Hepatol. 2007, 5, 496–501. [Google Scholar] [CrossRef]
- Asfari, M.M.; Sarmini, M.T.; Baidoun, F.; Al-Khadra, Y.; Ezaizi, Y.; Dasarathy, S.; McCullough, A. Association of non-alcoholic fatty liver disease and polycystic ovarian syndrome. BMJ Open Gastroenterol. 2020, 7, e000352. [Google Scholar] [CrossRef]
- Yao, K.; Bian, C.; Zhao, X. Association of polycystic ovary syndrome with metabolic syndrome and gestational diabetes: Aggravated complication of pregnancy. Exp. Ther. Med. 2017, 14, 1271–1276. [Google Scholar] [CrossRef] [Green Version]
- Manneras, L.; Cajander, S.; Holmäng, A.; Seleskovic, Z.; Lystig, T.; Lönn, M.; Stener-Victorin, E. A new rat model exhibiting both ovarian and metabolic characteristics of polycystic ovary syndrome. Endocrinology 2007, 148, 3781–3791. [Google Scholar] [CrossRef] [Green Version]
- Scicchitano, P.; Dentamaro, I.; Carbonara, R.; Bulzis, G.; Dachille, A.; Caputo, P.; Riccardi, R.; Locorotondo, M.; Mandurino, C.; Ciccone, M.M. Cardiovascular risk in women with PCOS. Int. J. Endocrinol. Metab. 2012, 10, 611. [Google Scholar] [CrossRef] [Green Version]
- Osibogun, O.; Ogunmoroti, O.; Michos, E.D. Polycystic ovary syndrome and cardiometabolic risk: Opportunities for cardiovascular disease prevention. Trends Cardiovasc. Med. 2020, 30, 399–404. [Google Scholar] [CrossRef] [PubMed]
- Sirmans, S.M.; Pate, K.A. Epidemiology, diagnosis, and management of polycystic ovary syndrome. Clin. Epidemiol. 2014, 6, 1. [Google Scholar] [CrossRef] [PubMed]
- Zhao, L.; Zhu, Z.; Lou, H.; Zhu, G.; Huang, W.; Zhang, S.; Liu, F. Polycystic ovary syndrome (PCOS) and the risk of coronary heart disease (CHD): A meta-analysis. Oncotarget 2016, 7, 33715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Y.; Wang, X.; Jiang, Y.; Ma, H.; Chen, L.; Lai, C.; Peng, C.; He, C.; Sun, C. Association between polycystic ovary syndrome and the risk of stroke and all-cause mortality: Insights from a meta-analysis. Gynecol. Endocrinol. 2017, 33, 904–910. [Google Scholar] [CrossRef]
- Wild, R.A.; Rizzo, M.; Clifton, S.; Carmina, E. Lipid levels in polycystic ovary syndrome: Systematic review and meta-analysis. Fertil. Steril. 2011, 95, 1073–1079.e1011. [Google Scholar] [CrossRef] [PubMed]
- Diamanti-Kandarakis, E.; Papavassiliou, A.G.; Kandarakis, S.A.; Chrousos, G.P. Pathophysiology and types of dyslipidemia in PCOS. Trends Endocrinol. Metab. 2007, 18, 280–285. [Google Scholar] [CrossRef] [PubMed]
- Ollila, M.-M.E.; Kaikkonen, K.; Järvelin, M.-R.; Huikuri, H.V.; Tapanainen, J.S.; Franks, S.; Piltonen, T.T.; Morin-Papunen, L. Self-reported polycystic ovary syndrome is associated with hypertension: A northern Finland birth cohort 1966 study. J. Clin. Endocrinol. Metab. 2019, 104, 1221–1231. [Google Scholar] [CrossRef]
- Hoeger, K.M.; Dokras, A.; Piltonen, T. Update on PCOS: Consequences, challenges, and guiding treatment. J. Clin. Endocrinol. Metab. 2021, 106, e1071–e1083. [Google Scholar] [CrossRef]
- Meyer, M.L.; Malek, A.M.; Wild, R.A.; Korytkowski, M.T.; Talbott, E.O. Carotid artery intima-media thickness in polycystic ovary syndrome: A systematic review and meta-analysis. Hum. Reprod. Update 2012, 18, 112–126. [Google Scholar] [CrossRef]
- Gibson-Helm, M.; Teede, H.; Dunaif, A.; Dokras, A. Delayed diagnosis and a lack of information associated with dissatisfaction in women with polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 2017, 102, 604–612. [Google Scholar] [CrossRef] [Green Version]
- Yang, R.; Yang, S.; Li, R.; Liu, P.; Qiao, J.; Zhang, Y. Effects of hyperandrogenism on metabolic abnormalities in patients with polycystic ovary syndrome: A meta-analysis. Reprod. Biol. Endocrinol. 2016, 14, 67. [Google Scholar] [CrossRef] [Green Version]
- Fazleen, N.E.; Whittaker, M.; Mamun, A. Risk of metabolic syndrome in adolescents with polycystic ovarian syndrome: A systematic review and meta-analysis. Diabetes Metab. Syndr. Clin. Res. Rev. 2018, 12, 1083–1090. [Google Scholar] [CrossRef]
- Barry, J.A.; Azizia, M.M.; Hardiman, P.J. Risk of endometrial, ovarian and breast cancer in women with polycystic ovary syndrome: A systematic review and meta-analysis. Hum. Reprod. Update 2014, 20, 748–758. [Google Scholar] [CrossRef]
- Hung, J.-H.; Hu, L.-Y.; Tsai, S.-J.; Yang, A.C.; Huang, M.-W.; Chen, P.-M.; Wang, S.-L.; Lu, T.; Shen, C.-C. Risk of psychiatric disorders following polycystic ovary syndrome: A nationwide population-based cohort study. PLoS ONE 2014, 9, e97041. [Google Scholar] [CrossRef] [Green Version]
- Cooney, L.G.; Lee, I.; Sammel, M.D.; Dokras, A. High prevalence of moderate and severe depressive and anxiety symptoms in polycystic ovary syndrome: A systematic review and meta-analysis. Hum. Reprod. 2017, 32, 1075–1091. [Google Scholar] [CrossRef] [Green Version]
- Lee, I.; Cooney, L.G.; Saini, S.; Sammel, M.D.; Allison, K.C.; Dokras, A. Increased odds of disordered eating in polycystic ovary syndrome: A systematic review and meta-analysis. Eat. Weight Disord. Stud. Anorex. Bulim. Obes. Rev. 2019, 24, 787–797. [Google Scholar] [CrossRef]
- Alur-Gupta, S.; Chemerinski, A.; Liu, C.; Lipson, J.; Allison, K.; Sammel, M.D.; Dokras, A. Body-image distress is increased in women with polycystic ovary syndrome and mediates depression and anxiety. Fertil. Steril. 2019, 112, 930–938.e931. [Google Scholar] [CrossRef]
- Barnard, L.; Ferriday, D.; Guenther, N.; Strauss, B.; Balen, A.; Dye, L. Quality of life and psychological well being in polycystic ovary syndrome. Hum. Reprod. 2007, 22, 2279–2286. [Google Scholar] [CrossRef] [Green Version]
- Coffey, S.; Bano, G.; Mason, H.D. Health-related quality of life in women with polycystic ovary syndrome: A comparison with the general population using the Polycystic Ovary Syndrome Questionnaire (PCOSQ) and the Short Form-36 (SF-36). Gynecol. Endocrinol. 2006, 22, 80–86. [Google Scholar] [CrossRef]
- Karjula, S.; Morin-Papunen, L.; Franks, S.; Auvinen, J.; Järvelin, M.-R.; Tapanainen, J.S.; Jokelainen, J.; Miettunen, J.; Piltonen, T.T. Population-based data at ages 31 and 46 show decreased HRQoL and life satisfaction in women with PCOS symptoms. J. Clin. Endocrinol. Metab. 2020, 105, 1814–1826. [Google Scholar] [CrossRef]
- Ergen, K.; Yildiz, F.; Ozcan, M.; Cekmen, M.; Ta Utkan, T.; Karakoc, Y. Oxidative stress status, metabolic profile and cardiovascular risk factors in patients with polycystic ovary syndrome. Med. Sci. 2012, 1, 27–34. [Google Scholar]
- Rudnicka, E.; Suchta, K.; Grymowicz, M.; Calik-Ksepka, A.; Smolarczyk, K.; Duszewska, A.M.; Smolarczyk, R.; Meczekalski, B. Chronic low grade inflammation in pathogenesis of PCOS. Int. J. Mol. Sci. 2021, 22, 3789. [Google Scholar] [CrossRef] [PubMed]
- Vassalli, P. The pathophysiology of tumor necrosis factors. Annu. Rev. Immunol. 1992, 10, 411–452. [Google Scholar] [CrossRef] [PubMed]
- Zolti, M.; Meirom, R.; Shemesh, M.; Wollach, D.; Mashiach, S.; Shore, L.; Ben Rafael, Z. Granulosa cells as a source and target organ for tumor necrosis factor-α. FEBS Lett. 1990, 261, 253–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.J.; Brännström, M.; Robertson, S.A.; Norman, R.J. Tumor necrosis factor α in the human ovary: Presence in follicular fluid and effects on cell proliferation and prostaglandin production. Fertil. Steril. 1992, 58, 934–940. [Google Scholar] [CrossRef]
- Veldhuis, J.D.; Garmey, J.C.; Urban, R.J.; Demers, L.M.; Aggarwal, B.B. Ovarian actions of tumor necrosis factor-α (TNFα): Pleiotropic effects of TNFα on differentiated functions of untransformed swine granulosa cells. Endocrinology 1991, 129, 641–648. [Google Scholar] [CrossRef]
- Roby, K.; Terranova, P. Effects of tumor necrosis factor-α in vitro on steroidogenesis of healthy and atretic follicles of the rat: Theca as a target. Endocrinology 1990, 126, 2711–2718. [Google Scholar] [CrossRef]
- Brannstrom, M.; Bonello, N.; Wang, L.J.; Norman, R.J. Effects of tumour necrosis factor alpha (TNF alpha) on ovulation in the rat ovary. Reprod. Fertil. Dev. 1995, 7, 67–73. [Google Scholar] [CrossRef]
- Hotamisligil, G.S.; Budavari, A.; Murray, D.; Spiegelman, B.M. Reduced tyrosine kinase activity of the insulin receptor in obesity-diabetes. Central role of tumor necrosis factor-alpha. J. Clin. Investig. 1994, 94, 1543–1549. [Google Scholar] [CrossRef]
- Stephens, J.M.; Pekala, P. Transcriptional repression of the GLUT4 and C/EBP genes in 3T3-L1 adipocytes by tumor necrosis factor-alpha. J. Biol. Chem. 1991, 266, 21839–21845. [Google Scholar] [CrossRef]
- Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
- Nilsson, M.B.; Langley, R.R.; Fidler, I.J. Interleukin-6, secreted by human ovarian carcinoma cells, is a potent proangiogenic cytokine. Cancer Res. 2005, 65, 10794–10800. [Google Scholar] [CrossRef] [Green Version]
- Adashi, E.Y. The potential relevance of cytokines to ovarian physiology: The emerging role of resident ovarian cells of the white blood cell series. Endocr. Rev. 1990, 11, 454–464. [Google Scholar] [CrossRef]
- Łukaszewicz, M.; Mroczko, B.; Szmitkowski, M. Clinical significance of interleukin-6 (IL-6) as a prognostic factor of cancer disease. Pol. Arch. Med. Wewn. 2007, 117, 247–251. [Google Scholar] [CrossRef] [Green Version]
- Xie, K. Interleukin-8 and human cancer biology. Cytokine Growth Factor Rev. 2001, 12, 375–391. [Google Scholar] [CrossRef]
- Arici, A.; Oral, E.; Bukulmez, O.; Buradagunta, S.; Engin, O.; Olive, D.L. Interleukin-8 expression and modulation in human preovulatory follicles and ovarian cells. Endocrinology 1996, 137, 3762–3769. [Google Scholar] [CrossRef] [Green Version]
- Rizk, B.; Aboulghar, M.; Smitz, J.; Ron-El, R. The role of vascular endothelial growth factor and interleukins in the pathogenesis of severe ovarian hyperstimulation syndrome. Hum. Reprod. Update 1997, 3, 255–266. [Google Scholar] [CrossRef] [Green Version]
- Chang, R.J.; Gougeon, A.; Erickson, G.F. Evidence for a neutrophil–interleukin-8 system in human folliculogenesis. Am. J. Obstet. Gynecol. 1998, 178, 650–657. [Google Scholar] [CrossRef]
- Moore, K.W.; de Waal Malefyt, R.; Coffman, R.L.; O’Garra, A. Interleukin-10 and the interleukin-10 receptor. Annu. Rev. Immunol. 2001, 19, 683. [Google Scholar] [CrossRef]
- Fiorentino, D.F.; Zlotnik, A.; Mosmann, T.R.; Howard, M.; O’Garra, A. IL-10 inhibits cytokine production by activated macrophages. J. Immunol. 1991, 147, 3815–3822. [Google Scholar] [CrossRef]
- Hashii, K.; Fujiwara, H.; Yoshioka, S.; Kataoka, N.; Yamada, S.; Hirano, T.; Mori, T.; Fujii, S.; Maeda, M. Peripheral blood mononuclear cells stimulate progesterone production by luteal cells derived from pregnant and non-pregnant women: Possible involvement of interleukin-4 and interleukin-10 in corpus luteum function and differentiation. Hum. Reprod. 1998, 13, 2738–2744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scarpelli, D.; Cardellini, M.; Andreozzi, F.; Laratta, E.; Hribal, M.L.; Marini, M.A.; Tassi, V.; Lauro, R.; Perticone, F.; Sesti, G. Variants of the interleukin-10 promoter gene are associated with obesity and insulin resistance but not type 2 diabetes in Caucasian Italian subjects. Diabetes 2006, 55, 1529–1533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vural, P.; Değirmencioğlu, S.; Saral, N.Y.; Akgül, C. Tumor necrosis factor α (−308), interleukin-6 (−174) and interleukin-10 (−1082) gene polymorphisms in polycystic ovary syndrome. Eur. J. Obstet. Gynecol. Reprod. Biol. Endocrinol. 2010, 150, 61–65. [Google Scholar] [CrossRef] [PubMed]
- Sirotkin, A.V. Cytokines: Signalling molecules controlling ovarian functions. Int. J. Biochem. Cell Biol. Int. 2011, 43, 857–861. [Google Scholar] [CrossRef] [PubMed]
- Vgontzas, A.N.; Bixler, E.; Chrousos, G. Metabolic disturbances in obesity versus sleep apnoea: The importance of visceral obesity and insulin resistance. J. Intern. Med. 2003, 254, 32–44. [Google Scholar] [CrossRef]
- González, F. Inflammation in polycystic ovary syndrome: Underpinning of insulin resistance and ovarian dysfunction. Steroids 2012, 77, 300–305. [Google Scholar] [CrossRef] [Green Version]
- Osborn, O.; Olefsky, J.M. The cellular and signaling networks linking the immune system and metabolism in disease. Nat. Med. 2012, 18, 363–374. [Google Scholar] [CrossRef]
- Deligeoroglou, E.; Vrachnis, N.; Athanasopoulos, N.; Iliodromiti, Z.; Sifakis, S.; Iliodromiti, S.; Siristatidis, C.; Creatsas, G. Mediators of chronic inflammation in polycystic ovarian syndrome. Gynecol. Endocrinol. 2012, 28, 974–978. [Google Scholar] [CrossRef]
- Kaya, C.; Pabuccu, R.; Berker, B.; Satıroglu, H. Plasma interleukin-18 levels are increased in the polycystic ovary syndrome: Relationship of carotid intima-media wall thickness and cardiovascular risk factors. Fertil. Steril. 2010, 93, 1200–1207. [Google Scholar] [CrossRef]
- Escobar-Morreale, H.c.F.; Botella-Carretero, J.I.; Villuendas, G.; Sancho, J.; San Millán, J.L. Serum interleukin-18 concentrations are increased in the polycystic ovary syndrome: Relationship to insulin resistance and to obesity. J. Clin. Endocrinol. Metab. 2004, 89, 806–811. [Google Scholar] [CrossRef] [Green Version]
- Toulis, K.A.; Goulis, D.G.; Mintziori, G.; Kintiraki, E.; Eukarpidis, E.; Mouratoglou, S.-A.; Pavlaki, A.; Stergianos, S.; Poulasouchidou, M.; Tzellos, T.G. Meta-analysis of cardiovascular disease risk markers in women with polycystic ovary syndrome. Hum. Reprod. Update 2011, 17, 741–760. [Google Scholar] [CrossRef] [Green Version]
- Zangeneh, F.Z.; Naghizadeh, M.M.; Masoumi, M. Polycystic ovary syndrome and circulating inflammatory markers. Int. J. Reprod. BioMed. 2017, 15, 375. [Google Scholar] [CrossRef] [Green Version]
- Sun, Q.; Li, J.; Gao, F. New insights into insulin: The anti-inflammatory effect and its clinical relevance. World J. Diabetes 2014, 5, 89. [Google Scholar] [CrossRef]
- Woodward, E.A.; Prêle, C.M.; Nicholson, S.E.; Kolesnik, T.B.; Hart, P.H. The anti-inflammatory effects of interleukin-4 are not mediated by suppressor of cytokine signalling-1 (SOCS1). Immunology 2010, 131, 118–127. [Google Scholar] [CrossRef]
- Banchereau, J.; Pascual, V.; O’garra, A. From IL-2 to IL-37: The expanding spectrum of anti-inflammatory cytokines. Nat. Immunol. 2012, 13, 925–931. [Google Scholar] [CrossRef]
- Tosatti, J.A.; Sóter, M.O.; Ferreira, C.N.; de FO Silva, I.; Cândido, A.L.; Sousa, M.O.; Reis, F.M.; Gomes, K.B. The hallmark of pro-and anti-inflammatory cytokine ratios in women with polycystic ovary syndrome. Cytokine Growth Factor Rev. 2020, 134, 155187. [Google Scholar] [CrossRef]
- Phelan, N.; O’Connor, A.; Kyaw Tun, T.; Correia, N.; Boran, G.; Roche, H.; Gibney, J. Leucocytosis in women with polycystic ovary syndrome (PCOS) is incompletely explained by obesity and insulin resistance. Clin. Endocrinol. 2013, 78, 107–113. [Google Scholar] [CrossRef]
- Wiernik, P.H. Androgen therapy for acute myeloid and hairy cell leukemia. Curr. Treat. Options Oncol. 2018, 19, 4. [Google Scholar] [CrossRef]
- Covington, J.D.; Tam, C.S.; Pasarica, M.; Redman, L.M. Higher circulating leukocytes in women with PCOS is reversed by aerobic exercise. Biochimie 2016, 124, 27–33. [Google Scholar] [CrossRef] [Green Version]
- Aboeldalyl, S.; James, C.; Seyam, E.; Ibrahim, E.M.; Shawki, H.E.-D.; Amer, S. The role of chronic inflammation in polycystic ovarian syndrome—A systematic review and meta-analysis. Int. J. Mol. Sci. 2021, 22, 2734. [Google Scholar] [CrossRef]
- Barvitenko, N.; Adragna, N.; Weber, R. Erythrocyte signal transduction pathways, their oxygenation dependence and functional significance. Cell. Physiol. Biochem. 2005, 15, 001–018. [Google Scholar] [CrossRef] [PubMed]
- Taylor, S.; Dirir, O.; Zamanian, R.T.; Rabinovitch, M.; Thompson, A.R. The role of neutrophils and neutrophil elastase in pulmonary arterial hypertension. Front. Med. 2018, 5, 217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Almaeen, A.H.; Alduraywish, A.A.; Nabi, M.; Shah, N.N.; Shaik, R.; Tantry, B.A. Quantitative Changes in White Blood Cells: Correlation with the Hallmarks of Polycystic Ovary Syndrome. Medicina 2022, 58, 535. [Google Scholar] [CrossRef] [PubMed]
- Çakıroğlu, Y.; Vural, F.; Vural, B. The inflammatory markers in polycystic ovary syndrome: Association with obesity and IVF outcomes. J. Endocrinol. Investig. 2016, 39, 899–907. [Google Scholar] [CrossRef] [PubMed]
- Yilmaz, M.; Duran, C.; Basaran, M. The mean platelet volume and neutrophil to lymphocyte ratio in obese and lean patients with polycystic ovary syndrome. J. Endocrinol. Investig. 2016, 39, 45–53. [Google Scholar] [CrossRef]
- Papalou, O.; Livadas, S.; Karachalios, A.; Tolia, N.; Kokkoris, P.; Tripolitakis, K.; Diamanti-Kandarakis, E. White blood cells levels and PCOS: Direct and indirect relationship with obesity and insulin resistance, but not with hyperandogenemia. Hormones 2015, 14, 91–100. [Google Scholar] [CrossRef]
- Shamasbi, S.G.; Ghanbari-Homayi, S.; Mirghafourvand, M. The effect of probiotics, prebiotics, and synbiotics on hormonal and inflammatory indices in women with polycystic ovary syndrome: A systematic review and meta-analysis. Eur. J. Nutr. 2020, 59, 433–450. [Google Scholar] [CrossRef]
- Cozzolino, M.; Vitagliano, A.; Pellegrini, L.; Chiurazzi, M.; Andriasani, A.; Ambrosini, G.; Garrido, N. Therapy with probiotics and synbiotics for polycystic ovarian syndrome: A systematic review and meta-analysis. Eur. J. Nutr. 2020, 59, 2841–2856. [Google Scholar] [CrossRef]
- Xu, L.-H.; Zhang, F. Meta-analysis of the endocrine and metabolic effects of probiotics on polycystic ovary syndrome. Res. Sq. 2022, preprint. [Google Scholar] [CrossRef]
- Ahmadi, S.; Jamilian, M.; Karamali, M.; Tajabadi-Ebrahimi, M.; Jafari, P.; Taghizadeh, M.; Memarzadeh, M.R.; Asemi, Z. Probiotic supplementation and the effects on weight loss, glycaemia and lipid profiles in women with polycystic ovary syndrome: A randomized, double-blind, placebo-controlled trial. Hum. Fertil. 2017, 20, 254–261. [Google Scholar] [CrossRef]
- Shoaei, T.; Heidari-Beni, M.; Tehrani, H.G. Effects of probiotic supplementation on pancreatic β-cell function and c-reactive protein in women with polycystic ovary syndrome: A randomized double-blind placebo-controlled clinical trial. Int. J. Prev. Med. 2015, 6, 27. [Google Scholar] [CrossRef]
- Rashad, N.M.; El-Shal, A.S.; Amin, A.I.; Soliman, M.H. Effects of probiotics supplementation on macrophage migration inhibitory factor and clinical laboratory feature of polycystic ovary syndrome. J. Funct. Foods 2017, 36, 317–324. [Google Scholar] [CrossRef]
- Jamilian, M.; Mansury, S.; Bahmani, F.; Heidar, Z.; Amirani, E.; Asemi, Z. The effects of probiotic and selenium co-supplementation on parameters of mental health, hormonal profiles, and biomarkers of inflammation and oxidative stress in women with polycystic ovary syndrome. J. Ovarian Res. 2018, 11, 80. [Google Scholar] [CrossRef]
- Ghanei, N.; Rezaei, N.; Amiri, G.A.; Zayeri, F.; Makki, G.; Nasseri, E. The probiotic supplementation reduced inflammation in polycystic ovary syndrome: A randomized, double-blind, placebo-controlled trial. J. Funct. Foods 2018, 42, 306–311. [Google Scholar] [CrossRef]
- Zhang, P.; Feng, Y.; Li, L.; Ge, W.; Yu, S.; Hao, Y.; Shen, W.; Han, X.; Ma, D.; Yin, S.; et al. Improvement in sperm quality and spermatogenesis following faecal microbiota transplantation from alginate oligosaccharide dosed mice. Gut 2021, 70, 222–225. [Google Scholar] [CrossRef] [Green Version]
- Quaranta, G.; Sanguinetti, M.; Masucci, L. Fecal Microbiota Transplantation: A Potential Tool for Treatment of Human Female Reproductive Tract Diseases. Front. Immunol. 2019, 10, 2653. [Google Scholar] [CrossRef]
- Gao, X.; Liu, K.; Huang, F.; Zhang, D.; Guo, X.; Wang, M.; Liu, B. Positive and negative regulation of insulin action by genistein in the endothelium. J. Nutr. Biochem. 2013, 24, 222–230. [Google Scholar] [CrossRef]
- Yanagisawa, M.; Sugiya, M.; Iijima, H.; Nakagome, I.; Hirono, S.; Tsuda, T. Genistein and daidzein, typical soy isoflavones, inhibit TNF-α-mediated downregulation of adiponectin expression via different mechanisms in 3T3-L1 adipocytes. Mol. Nutr. Food Res. 2012, 56, 1783–1793. [Google Scholar] [CrossRef]
- Khani, B.; Mehrabian, F.; Khalesi, E.; Eshraghi, A. Effect of soy phytoestrogen on metabolic and hormonal disturbance of women with polycystic ovary syndrome. J. Res. Med. Sci. 2011, 16, 297. [Google Scholar]
- Jamilian, M.; Asemi, Z. The effects of soy isoflavones on metabolic status of patients with polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 2016, 101, 3386–3394. [Google Scholar] [CrossRef] [Green Version]
- Rajaei, S.; Alihemmati, A.; Abedelahi, A. Antioxidant effect of genistein on ovarian tissue morphology, oxidant and antioxidant activity in rats with induced polycystic ovary syndrome. Int. J. Reprod. BioMed. 2019, 17, 11. [Google Scholar] [CrossRef] [PubMed]
- Alivandi Farkhad, S.; Khazali, H. Therapeutic effects of isoflavone-aglycone fraction from soybean (Glycine max L. Merrill) in rats with estradiol valerate-induced polycystic ovary syndrome as an inflammatory state. Gynecol. Endocrinol. 2019, 35, 1078–1083. [Google Scholar] [CrossRef] [PubMed]
- Zhang, T.; Chi, X. Estrogenic properties of genistein acting on FSHR and LHR in rats with PCOS. Pol. J. Vet. Sci. 2019. [Google Scholar] [CrossRef]
- Haudum, C.; Lindheim, L.; Ascani, A.; Trummer, C.; Horvath, A.; Münzker, J.; Obermayer-Pietsch, B. Impact of Short-Term Isoflavone Intervention in Polycystic Ovary Syndrome (PCOS) Patients on Microbiota Composition and Metagenomics. Nutrients 2020, 12, 1622. [Google Scholar] [CrossRef] [PubMed]
- Franke, A.A.; Lai, J.F.; Halm, B.M. Absorption, distribution, metabolism, and excretion of isoflavonoids after soy intake. Arch. Biochem. Biophys. 2014, 559, 24–28. [Google Scholar] [CrossRef] [Green Version]
- Patisaul, H.B.; Mabrey, N.; Adewale, H.B.; Sullivan, A.W. Soy but not bisphenol A (BPA) induces hallmarks of polycystic ovary syndrome (PCOS) and related metabolic co-morbidities in rats. Reprod. Toxicol. 2014, 49, 209–218. [Google Scholar] [CrossRef] [Green Version]
- Romualdi, D.; Costantini, B.; Campagna, G.; Lanzone, A.; Guido, M. Is there a role for soy isoflavones in the therapeutic approach to polycystic ovary syndrome? Results from a pilot study. Fertil. Steril. 2008, 90, 1826–1833. [Google Scholar] [CrossRef]
- Balasubramanian, M.M.; Palayyan, M.; Rajeswary, H. Effect of ethanolic seed extract of Caesalpinia bonducella on hormones in mifepristone induced PCOS rats. J. Appl. Pharm. Sci. 2020, 10, 072–076. [Google Scholar]
- Salunke, K.R.; Ahmed, R.N.; Marigoudar, S.R. Effect of graded doses of Caesalpinia bonducella seed extract on ovary and uterus in albino rats. J. Basic Clin. Physiol. Pharmacol. 2011, 22, 49–53. [Google Scholar] [CrossRef]
- Kandasamy, V.; Balasundaram, U. Caesalpinia bonduc (L.) Roxb. as a promising source of pharmacological compounds to treat Poly Cystic Ovary Syndrome (PCOS): A review. J. Ethnopharmacol. 2021, 279, 114375. [Google Scholar] [CrossRef]
- Pascoe, K.O.; Burke, B.A.; Chan, W.R. Caesalpin F: A New Furanoditerpene from Caesalpinia bonducella. J. Nat. Prod. 1986, 49, 913–915. [Google Scholar] [CrossRef]
- Balasubramanian, M.; Palayyan, M.; Rajeswary, H. Effect of ethanolic seed extract of Caesalpinia bonducella on Mifepristone induced PCOS rats. Biomedicine 2022, 39, 274–281. [Google Scholar] [CrossRef]
- Sivalingam, V.N.; Myers, J.; Nicholas, S.; Balen, A.H.; Crosbie, E.J. Metformin in reproductive health, pregnancy and gynaecological cancer: Established and emerging indications. Hum. Reprod. Update 2014, 20, 853–868. [Google Scholar] [CrossRef]
- Jensterle, M.; Kravos, N.A.; Ferjan, S.; Goricar, K.; Dolzan, V.; Janez, A. Long-term efficacy of metformin in overweight-obese PCOS: Longitudinal follow-up of retrospective cohort. Endocr. Connect. 2020, 9, 44–54. [Google Scholar] [CrossRef] [Green Version]
- Barbieri, R.L. Metformin for the treatment of polycystic ovary syndrome. Obstet. Gynecol. 2003, 101, 785–793. [Google Scholar] [CrossRef]
- Genazzani, A.D.; Ricchieri, F.; Lanzoni, C. Use of Metformin in the Treatment of Polycystic Ovary Syndrome. Women’s Health 2010, 6, 577–593. [Google Scholar] [CrossRef]
- Tang, T.; Lord, J.M.; Norman, R.J.; Yasmin, E.; Balen, A.H. Insulin-sensitising drugs (metformin, rosiglitazone, pioglitazone, D-chiro-inositol) for women with polycystic ovary syndrome, oligo amenorrhoea and subfertility. Cochrane Database Syst. Rev. 2012, 5, CD003053. [Google Scholar] [CrossRef]
- Spiegelman, B.M. PPAR-gamma: Adipogenic regulator and thiazolidinedione receptor. Diabetes 1998, 47, 507–514. [Google Scholar] [CrossRef]
- Anam, A.K.; Inzucchi, S.E. Newer Glucose-Lowering Medications and Potential Role in Metabolic Management of PCOS. In Polycystic Ovary Syndrome: Current and Emerging Concepts; Pal, L., Seifer, D.B., Eds.; Springer International Publishing: Cham, Switzerland, 2022; pp. 527–553. [Google Scholar] [CrossRef]
- Vella, A. Mechanism of action of DPP-4 inhibitors—New insights. J. Clin. Endocrinol. Metab. 2012, 97, 2626–2628. [Google Scholar] [CrossRef] [Green Version]
- Thornberry, N.A.; Gallwitz, B. Mechanism of action of inhibitors of dipeptidyl-peptidase-4 (DPP-4). Best Pract. Res. Clin. Endocrinol. Metab. 2009, 23, 479–486. [Google Scholar] [CrossRef]
- Cefalu, W.T. The physiologic role of incretin hormones: Clinical applications. J. Am. Osteopath. Assoc. 2010, 110, S8–S14. [Google Scholar] [PubMed]
- He, Y.; Wang, Q.; Li, X.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Lactic acid bacteria alleviate polycystic ovarian syndrome by regulating sex hormone related gut microbiota. Food Funct. 2020, 11, 5192–5204. [Google Scholar] [CrossRef] [PubMed]
- Gholizadeh Shamasbi, S.; Dehgan, P.; Mohammad-Alizadeh Charandabi, S.; Aliasgarzadeh, A.; Mirghafourvand, M. The effect of resistant dextrin as a prebiotic on metabolic parameters and androgen level in women with polycystic ovarian syndrome: A randomized, triple-blind, controlled, clinical trial. Eur. J. Nutr. 2019, 58, 629–640. [Google Scholar] [CrossRef] [PubMed]
- Glueck, C.J.; Wang, P.; Kobayashi, S.; Phillips, H.; Sieve-Smith, L. Metformin therapy throughout pregnancy reduces the development of gestational diabetes in women with polycystic ovary syndrome. Fertil. Steril. 2002, 77, 520–525. [Google Scholar] [CrossRef]
- Muscelli, E.; Casolaro, A.; Gastaldelli, A.; Mari, A.; Seghieri, G.; Astiarraga, B.; Chen, Y.; Alba, M.; Holst, J.; Ferrannini, E. Mechanisms for the antihyperglycemic effect of sitagliptin in patients with type 2 diabetes. J. Clin. Endocrinol. Metab. 2012, 97, 2818–2826. [Google Scholar] [CrossRef] [Green Version]
- Naka, K.K.; Kalantaridou, S.N.; Kravariti, M.; Bechlioulis, A.; Kazakos, N.; Calis, K.A.; Makrigiannakis, A.; Katsouras, C.S.; Chrousos, G.P.; Tsatsoulis, A.; et al. Effect of the insulin sensitizers metformin and pioglitazone on endothelial function in young women with polycystic ovary syndrome: A prospective randomized study. Fertil. Steril. 2011, 95, 203–209. [Google Scholar] [CrossRef]
- Baranowska-Bik, A. Therapy of obesity in women with PCOS using GLP-1 analogues—Benefits and limitations [Terapia otyłości u kobiet z PCOS przy zastosowaniu analogów GLP-1—Korzyści i ograniczenia]. Endokrynol. Pol. 2022, 73, 627–643. [Google Scholar] [CrossRef]
Effect | Microorganism | Changes | References |
---|---|---|---|
Increase | Escherichia and Shigella | Alteration in SCFAs production | [73] |
Decrease | Lactobacilli and Bifidobacterium | Promote nutrition absorption and enhance immunity | [54] |
Increase | Bacteroides vulgatus | Reduced levels of glycodeoxycholic and tauroursodeoxycholic acid | [75] |
Increase | Prevotella | Detrimental inflammatory effect on the host | [76] |
Decrease | Prevotella | Loss in anti-inflammatory metabolites | [77] |
Sample | Effect | Inflammatory Markers | Observation | Reference |
---|---|---|---|---|
Serum and endometrial tissue | Increase | TNFα | PCOS aggravated by obesity condition leads to an inflammatory state | [154] |
Serum | Increase | IL-6 | Raised IL-6 levels are related to androgens and IR but not to BMI | [155] |
Adipose tissue of rat | Increase | IL-6 | When treated with resveratrol, IL-6 mRNA expression decreased compared to normal treatment | [156] |
Serum and follicular fluid | Increase | IL-8 | In GCs with an increase in BMI, increased IL-8 mRNA expression but gradually decreased serum concentration | [157] |
Follicular fluid | Decrease | IL-10 | Reduced IL-10 leads to oxidative stress in PCOS, which in turn leads to inflammatory and androgen synthesis | [158] |
Serum and pooled follicular fluid | Increase | IL-18 | Elevated IL-18 levels affect the ovary leading to folliculogenesis disruption | [159] |
Serum | Increase | CRP | Obese PCOS females are at IR risk where oxidative stress does not cause obesity | [160] |
Therapeutic Options | Mechanism | Experimental Model | Dosage | Effects | Reference |
---|---|---|---|---|---|
Probiotics | Regulate sex hormone-related microbes | Sprague Dawley rats | 1 × 109 CFU bacteria for 28 days | Reduced MDA and FAI; increased NO and SHBG; increased TAC and GSH levels | [307,343] |
Prebiotics | Improve inflammation, antioxidant activity | Humans | 20 g for 3 months | Reduced expression of inflammatory and oxidative stress markers | [307,344] |
Fecal microbiota transplantation | Restores gut flora alterations | Sprague-Dawley rats | 2 × 109 fecal microbiota for 14 days | Decreased androgen level, increased estrogen level; improved ovarian disorder and estrus cycles | [83] |
Isoflavone interventions | Have estrogen-modulating, antioxidant, and anti-inflammatory activities | Human | 200 mL soy drink; twice a day for 3 days | Improved glucose homeostasis, stool metagenomic pathways, microbial α -diversity | [325] |
Caesalpinia bonducella | Induces ovulation | Wistar strain adult albino female rats | 200–400 mg/kg for 28 days | Ameliorated HI, insulin resistance, and HA | [328] |
Metformin treatment | Induces high glucose uptake | Humans | 2.55 g/d throughout pregnancy | Reduced gestational diabetes; IR; insulin secretion | [345] |
DPP-4 inhibitor: sitagliptin | Increases incretin hormone, GLP1, and GIP | Humans | 100 mg/d for 6 weeks | Improved insulin sensitivity; β-cell glucose sensing; lowered oral glucose, glucagon response, and postprandial endogenous glucose release | [346] |
Pioglitazone | Improves IR | Humans | 30 mg/day for 6 months | Improved endothelial function; decreased cardiovascular risk | [347] |
GLP-1 receptor agonists | Reduces BMI; improves IR; decreases abdominal circumferences | Humans | 1.2 mg of liraglutide once daily | Improved IR; decreases HA. | [348] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mukherjee, A.G.; Wanjari, U.R.; Kannampuzha, S.; Murali, R.; Namachivayam, A.; Ganesan, R.; Dey, A.; Babu, A.; Renu, K.; Vellingiri, B.; et al. The Implication of Mechanistic Approaches and the Role of the Microbiome in Polycystic Ovary Syndrome (PCOS): A Review. Metabolites 2023, 13, 129. https://doi.org/10.3390/metabo13010129
Mukherjee AG, Wanjari UR, Kannampuzha S, Murali R, Namachivayam A, Ganesan R, Dey A, Babu A, Renu K, Vellingiri B, et al. The Implication of Mechanistic Approaches and the Role of the Microbiome in Polycystic Ovary Syndrome (PCOS): A Review. Metabolites. 2023; 13(1):129. https://doi.org/10.3390/metabo13010129
Chicago/Turabian StyleMukherjee, Anirban Goutam, Uddesh Ramesh Wanjari, Sandra Kannampuzha, Reshma Murali, Arunraj Namachivayam, Raja Ganesan, Abhijit Dey, Achsha Babu, Kaviyarasi Renu, Balachandar Vellingiri, and et al. 2023. "The Implication of Mechanistic Approaches and the Role of the Microbiome in Polycystic Ovary Syndrome (PCOS): A Review" Metabolites 13, no. 1: 129. https://doi.org/10.3390/metabo13010129
APA StyleMukherjee, A. G., Wanjari, U. R., Kannampuzha, S., Murali, R., Namachivayam, A., Ganesan, R., Dey, A., Babu, A., Renu, K., Vellingiri, B., Ramanathan, G., Priya Doss C., G., Elsherbiny, N., Elsherbini, A. M., Alsamman, A. M., Zayed, H., & Gopalakrishnan, A. V. (2023). The Implication of Mechanistic Approaches and the Role of the Microbiome in Polycystic Ovary Syndrome (PCOS): A Review. Metabolites, 13(1), 129. https://doi.org/10.3390/metabo13010129