1
|
Müller TD, Adriaenssens A, Ahrén B, Blüher M, Birkenfeld AL, Campbell JE, Coghlan MP, D'Alessio D, Deacon CF, DelPrato S, Douros JD, Drucker DJ, Figueredo Burgos NS, Flatt PR, Finan B, Gimeno RE, Gribble FM, Hayes MR, Hölscher C, Holst JJ, Knerr PJ, Knop FK, Kusminski CM, Liskiewicz A, Mabilleau G, Mowery SA, Nauck MA, Novikoff A, Reimann F, Roberts AG, Rosenkilde MM, Samms RJ, Scherer PE, Seeley RJ, Sloop KW, Wolfrum C, Wootten D, DiMarchi RD, Tschöp MH. Glucose-dependent insulinotropic polypeptide (GIP). Mol Metab 2025; 95:102118. [PMID: 40024571 PMCID: PMC11931254 DOI: 10.1016/j.molmet.2025.102118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Glucose-dependent insulinotropic polypeptide (GIP) was the first incretin identified and plays an essential role in the maintenance of glucose tolerance in healthy humans. Until recently GIP had not been developed as a therapeutic and thus has been overshadowed by the other incretin, glucagon-like peptide 1 (GLP-1), which is the basis for several successful drugs to treat diabetes and obesity. However, there has been a rekindling of interest in GIP biology in recent years, in great part due to pharmacology demonstrating that both GIPR agonism and antagonism may be beneficial in treating obesity and diabetes. This apparent paradox has reinvigorated the field, led to new lines of investigation, and deeper understanding of GIP. SCOPE OF REVIEW In this review, we provide a detailed overview on the multifaceted nature of GIP biology and discuss the therapeutic implications of GIPR signal modification on various diseases. MAJOR CONCLUSIONS Following its classification as an incretin hormone, GIP has emerged as a pleiotropic hormone with a variety of metabolic effects outside the endocrine pancreas. The numerous beneficial effects of GIPR signal modification render the peptide an interesting candidate for the development of pharmacotherapies to treat obesity, diabetes, drug-induced nausea and both bone and neurodegenerative disorders.
Collapse
Affiliation(s)
- Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany; Walther-Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University Munich (LMU), Germany.
| | - Alice Adriaenssens
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Bo Ahrén
- Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Matthias Blüher
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen 72076, Germany; Institute of Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich, Tübingen, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Department of Medicine, Division of Endocrinology, Duke University, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Matthew P Coghlan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - David D'Alessio
- Department of Medicine, Division of Endocrinology, Duke University, Durham, NC, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Carolyn F Deacon
- School of Biomedical Sciences, Ulster University, Coleraine, UK; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stefano DelPrato
- Interdisciplinary Research Center "Health Science", Sant'Anna School of Advanced Studies, Pisa, Italy
| | | | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, and the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Natalie S Figueredo Burgos
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Peter R Flatt
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Brian Finan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Ruth E Gimeno
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Fiona M Gribble
- Institute of Metabolic Science-Metabolic Research Laboratories & MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Matthew R Hayes
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christian Hölscher
- Neurodegeneration Research Group, Henan Academy of Innovations in Medical Science, Xinzheng, China
| | - Jens J Holst
- Department of Biomedical Sciences and the Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Patrick J Knerr
- Indianapolis Biosciences Research Institute, Indianapolis, IN, USA
| | - Filip K Knop
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christine M Kusminski
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arkadiusz Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany; Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Guillaume Mabilleau
- Univ Angers, Nantes Université, ONIRIS, Inserm, RMeS UMR 1229, Angers, France; CHU Angers, Departement de Pathologie Cellulaire et Tissulaire, Angers, France
| | | | - Michael A Nauck
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Aaron Novikoff
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany
| | - Frank Reimann
- Institute of Metabolic Science-Metabolic Research Laboratories & MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Anna G Roberts
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences University of Copenhagen, Copenhagen, Denmark
| | - Ricardo J Samms
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Philip E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kyle W Sloop
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Denise Wootten
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia; ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | | | - Matthias H Tschöp
- Helmholtz Munich, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
2
|
Parahuleva N, Mihaylova A, Harizanova S, Merdzhanova Y, Koleva M, Madzharov V, Strikova G, Uchikova E. The Anti-Mullerian Hormone as a Biomarker of Effectiveness of Metformin Hydrochloride Therapy in Polycystic Ovarian Syndrome and Insulin Resistance. Healthcare (Basel) 2025; 13:884. [PMID: 40281834 PMCID: PMC12027305 DOI: 10.3390/healthcare13080884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Among the therapeutic options available for managing PCOS, metformin improves insulin sensitivity, reduces androgen levels, and helps restore menstrual regularity and ovulation. While primarily used for its metabolic effects, metformin therapy may also influence reproductive parameters, including AMH levels, which are pivotal in improving ovarian function and predicting therapeutic outcomes in PCOS. The aim of this study was to search the scientific literature and analyze the correlation between AMH levels and metformin hydrochloride therapy in women with PCOS and IR. Methods: A systematic review of the scientific literature was conducted using the following keywords: polycystic ovarian syndrome, anti-Mullerian hormone, insulin resistance, metformin, treatment, biomarker, and metabolic syndrome. This review was aimed at investigating the potential of AMH as a biomarker of the effectiveness of metformin therapy in patients with PCOS and IR. Results: Metformin treatment in PCOS patients has shown significant reductions in serum AMH levels with prolonged therapy. As an insulin sensitizer, metformin improves insulin sensitivity, reduces hyperinsulinemia, and suppresses hyperandrogenism. This process inhibits the growth of antral follicles, which is reflected in decreased AMH levels. Conclusions: Reductions in AMH levels and improvements in insulin sensitivity can serve as indicators of treatment efficacy and enhancements in reproductive function for these patients. AMH could be considered a prognostic marker for evaluating the effectiveness of metformin therapy. A decrease in AMH levels following treatment may indicate improved ovarian function and a reduction in polycystic morphology. However, further research is necessary to confirm these findings and to determine the optimal dosages and duration of treatment.
Collapse
Affiliation(s)
- Nikoleta Parahuleva
- Department of Obstetrics and Gynecology, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (N.P.); (Y.M.); (E.U.)
| | - Anna Mihaylova
- Department of Health Care Management, Faculty of Public Health, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Stanislava Harizanova
- Department of Hygiene, Faculty of Public Health, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Yana Merdzhanova
- Department of Obstetrics and Gynecology, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (N.P.); (Y.M.); (E.U.)
| | - Mariya Koleva
- Department of General and Clinical Pathology, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Vasil Madzharov
- Department of Organization and Economics of Pharmacy, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Gergana Strikova
- Medical College, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Ekaterina Uchikova
- Department of Obstetrics and Gynecology, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (N.P.); (Y.M.); (E.U.)
| |
Collapse
|
3
|
Amini M, Bahmani F, Foroozanfard F, Vahedpoor Z, Ghaderi A, Taghizadeh M, Karbassizadeh H, Asemi Z. Retracted article: The effects of fish oil omega-3 fatty acid supplementation on mental health parameters and metabolic status of patients with polycystic ovary syndrome: a randomized, double-blind, placebo-controlled trial. J Psychosom Obstet Gynaecol 2024; 45:1508282. [PMID: 30230402 DOI: 10.1080/0167482x.2018.1508282] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/24/2018] [Accepted: 07/29/2018] [Indexed: 01/28/2023] Open
Abstract
We, the Editor and Publisher of the Journal of Psychosomatic Obstetrics & Gynecology have retracted the following article:Mehrdad Amini, Fereshteh Bahmani, Fatemeh Foroozanfard, Zahra Vahedpoor, Amir Ghaderi, Mohsen Taghizadeh, Hassan Karbassizadeh & Zatollah Asemi (2018), The effects of fish oil omega-3 fatty acid supplementation on mental health parameters and metabolic status of patients with polycystic ovary syndrome: a randomized, double-blind, placebo-controlled trial. Journal of Psychosomatic Obstetrics & Gynecology, DOI: 10.1080/0167482X.2018.1508282.Following publication, significant concerns were raised by a third party 1 about the integrity of the data and the reported findings in the article.When approached for an explanation, the authors and their institution have been cooperative in providing some responses and documents. To verify the reported findings, the article and the documents provided by the authors were further evaluated by the journal editorial team, and also sent for review by an external statistical reviewer.Both the journal editorial team and the external statistical reviewer were unable to confirm the integrity of the trial design and the main outcome of the external review was that the article's results and conclusions are unreliable. Therefore, as the editorial team no longer have confidence in the reported conclusions the decision has been made to retract the article.The authors listed in the publication have been informed. The authors do not agree with the retraction.We have been informed in our decision-making by our editorial policies and the COPE guidelines.The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as 'Retracted'.
Collapse
Affiliation(s)
- Mehrdad Amini
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Fereshteh Bahmani
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Foroozanfard
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Vahedpoor
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Ghaderi
- Department of Addiction studies, School of Medical, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
4
|
Yun C, Yan S, Liao B, Ding Y, Qi X, Zhao M, Wang K, Zhuo Y, Nie Q, Ye C, Xia P, Ma M, Li R, Jiang C, Qiao J, Pang Y. The microbial metabolite agmatine acts as an FXR agonist to promote polycystic ovary syndrome in female mice. Nat Metab 2024; 6:947-962. [PMID: 38769396 DOI: 10.1038/s42255-024-01041-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 04/02/2024] [Indexed: 05/22/2024]
Abstract
Polycystic ovary syndrome (PCOS), an endocrine disorder afflicting 6-20% of women of reproductive age globally, has been linked to alterations in the gut microbiome. We previously showed that in PCOS, elevation of Bacteroides vulgatus in the gut microbiome was associated with altered bile acid metabolism. Here we show that B. vulgatus also induces a PCOS-like phenotype in female mice via an alternate mechanism independent of bile acids. We find that B. vulgatus contributes to PCOS-like symptoms through its metabolite agmatine, which is derived from arginine by arginine decarboxylase. Mechanistically, agmatine activates the farnesoid X receptor (FXR) pathway to subsequently inhibit glucagon-like peptide-1 (GLP-1) secretion by L cells, which leads to insulin resistance and ovarian dysfunction. Critically, the GLP-1 receptor agonist liraglutide and the arginine decarboxylase inhibitor difluoromethylarginine ameliorate ovarian dysfunction in a PCOS-like mouse model. These findings reveal that agmatine-FXR-GLP-1 signalling contributes to ovarian dysfunction, presenting a potential therapeutic target for PCOS management.
Collapse
Affiliation(s)
- Chuyu Yun
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Sen Yan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Baoying Liao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yong Ding
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xinyu Qi
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yingying Zhuo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Qixing Nie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Chuan Ye
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Pengyan Xia
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing, China
| | - Ming Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Rong Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China.
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China.
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing, China.
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China.
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China.
| | - Yanli Pang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China.
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
5
|
Aukan MI, Coutinho S, Pedersen SA, Simpson MR, Martins C. Differences in gastrointestinal hormones and appetite ratings between individuals with and without obesity-A systematic review and meta-analysis. Obes Rev 2023; 24:e13531. [PMID: 36416279 PMCID: PMC10078575 DOI: 10.1111/obr.13531] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/26/2022] [Accepted: 10/27/2022] [Indexed: 11/24/2022]
Abstract
Determining if gastrointestinal (GI) hormone response to food intake differs between individuals with, and without, obesity may improve our understanding of obesity pathophysiology. A systematic review and meta-analysis of studies assessing the concentrations of GI hormones, as well as appetite ratings, following a test meal, in individuals with and without obesity was undertaken. Systematic searches were conducted in the databases MEDLINE, Embase, Cochrane Library, PsycINFO, Web of Science, and ClinicalTrials.gov. A total of 7514 unique articles were retrieved, 115 included in the systematic review, and 70 in the meta-analysis. The meta-analysis compared estimated standardized mean difference in GI hormones' concentration, as well as appetite ratings, between individuals with and without obesity. Basal and postprandial total ghrelin concentrations were lower in individuals with obesity compared with controls, and this was reflected by lower postprandial hunger ratings in the former. Individuals with obesity had a lower postprandial concentration of total peptide YY compared with controls, but no significant differences were found for glucagon-like peptide 1, cholecystokinin, or other appetite ratings. A large methodological and statistical heterogeneity among studies was found. More comprehensive studies are needed to understand if the differences observed are a cause or a consequence of obesity.
Collapse
Affiliation(s)
- Marthe Isaksen Aukan
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Centre of Obesity and Innovation (ObeCe), Clinic of Surgery, St. Olav University Hospital, Trondheim, Norway
| | - Silvia Coutinho
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Public Health Nutrition at the Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo (UiO), Oslo, Norway
| | - Sindre Andre Pedersen
- Library Section for Research Support, Data and Analysis, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway.,Clinical Research Unit Central Norway, St. Olavs Hospital, Trondheim, Norway
| | - Catia Martins
- Obesity Research Group, Department of Clinical and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Centre of Obesity and Innovation (ObeCe), Clinic of Surgery, St. Olav University Hospital, Trondheim, Norway.,Department of Nutrition Sciences, the University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| |
Collapse
|
6
|
Jensterle M, Herman R, Janež A. Therapeutic Potential of Glucagon-like Peptide-1 Agonists in Polycystic Ovary Syndrome: From Current Clinical Evidence to Future Perspectives. Biomedicines 2022; 10:1989. [PMID: 36009535 PMCID: PMC9405922 DOI: 10.3390/biomedicines10081989] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/30/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the continuous effort to understand the pathophysiology and determine potential therapeutic targets, PCOS treatment largely depends on lifestyle intervention and symptomatic management of individual signs and symptoms. International guidelines recognize the importance of weight reduction as a cornerstone for the achievement of better metabolic, reproductive, and cardiovascular outcomes in PCOS women who are overweight or obese. With its profound weight loss potential in patients with or without diabetes, the administration of GLP-1 receptor agonists has been investigated in overweight/obese women with PCOS in several single-center randomized control trials with considerable variation in the dosing regimen, follow-up duration, and outcome measurements over recent years. Most trials reported superior weight loss effects of GLP-1 receptor agonists compared to lifestyle changes or metformin, with additional metabolic, reproductive, and cardiovascular benefits in this population. However, their use is currently not widely accepted by the clinical community that treats this population. The major concern is how to balance the reproductive and metabolic treatment strategies since the use of GLP-1 receptor agonists requires effective contraception while on therapy and a washout period before pregnancy. Both approaches are not mutually exclusive, yet the best choice requires a careful assessment of the clinical context. Knowing a patient's individual circumstances, precise clinical sub-phenotyping, and regular monitoring are crucial components for the safe and effective use of these new tools. In the present narrative review, we explore the current clinical evidence and provide the future perspectives and challenges for their implementation in PCOS management.
Collapse
Affiliation(s)
- Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Rok Herman
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Andrej Janež
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
7
|
Amisi CA. Markers of insulin resistance in Polycystic ovary syndrome women: An update. World J Diabetes 2022; 13:129-149. [PMID: 35432749 PMCID: PMC8984569 DOI: 10.4239/wjd.v13.i3.129] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 09/14/2021] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders, affecting 5%-10% of women of reproductive age. The importance of this syndrome lies in the magnitude of associated comorbidities: infertility, metabolic dysfunction, cardiovascular disease (CVD), plus psychological and oncological complications. Insulin resistance (IR) is a prominent feature of PCOS with a prevalence of 35%-80%. Without adequate management, IR with compensatory hyperinsulinemia contributes directly to reproductive dysfunction in women with PCOS. Furthermore, epidemiological data shows compelling evidence that PCOS is associated with an increased risk of impaired glucose tolerance, gestational diabetes mellitus and type 2 diabetes. In addition, metabolic dysfunction leads to a risk for CVD that increases with aging in women with PCOS. Indeed, the severity of IR in women with PCOS is associated with the amount of abdominal obesity, even in lean women with PCOS. Given these drastic implications, it is important to diagnose and treat insulin resistance as early as possible. Many markers have been proposed. However, quantitative assessment of IR in clinical practice remains a major challenge. The gold standard method for assessing insulin sensitivity is the hyperinsulinemic euglycemic glucose clamp. However, it is not used routinely because of the complexity of its procedure. Consequently, there has been an urgent need for surrogate markers of IR that are more applicable in large population-based epidemiological investigations. Despite this, many of them are either difficult to apply in routine clinical practice or useless for women with PCOS. Considering this difficulty, there is still a need for an accurate marker for easy, early detection and assessment of IR in women with PCOS. This review highlights markers of IR already used in women with PCOS, including new markers recently reported in literature, and it establishes a new classification for these markers.
Collapse
Affiliation(s)
- Chantal Anifa Amisi
- Endocrinology and Diabetes Unit, Department of Medicine, Universita Campus Bio-medico di Rome, Rome 00128, Italy
| |
Collapse
|
8
|
Review of Novel Potential Insulin Resistance Biomarkers in PCOS Patients—The Debate Is Still Open. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19042099. [PMID: 35206286 PMCID: PMC8871992 DOI: 10.3390/ijerph19042099] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/27/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022]
Abstract
Research on proteins and peptides that play roles in metabolic regulation, which may be considered potential insulin resistance markers in some medical conditions, such as diabetes mellitus, obesity and polycystic ovarian syndrome (PCOS), has recently gained in interest. PCOS is a common endocrine disorder associated with hyperandrogenemia and failure of ovulation, which is often accompanied by metabolic abnormalities, including obesity, dyslipidemia, hyperinsulinemia, and insulin resistance. In this review, we focus on less commonly known peptides/proteins and investigate their role as potential biomarkers for insulin resistance in females affected by PCOS. We summarize studies comparing the serum fasting concentration of particular agents in PCOS individuals and healthy controls. Based on our analysis, we propose that, in the majority of studies, the levels of nesfastin-1, myonectin, omentin, neudesin were decreased in PCOS patients, while the levels of the other considered agents (e.g., preptin, gremlin-1, neuregulin-4, xenopsin-related peptide, xenin-25, and galectin-3) were increased. However, there also exist studies presenting contrary results; in particular, most data existing for lipocalin-2 are inconsistent. Therefore, further research is required to confirm those hypotheses, as well as to elucidate the involvement of these factors in PCOS-related metabolic complications.
Collapse
|
9
|
Polat S, Şimşek Y. Plasminogenactivator inhibitor-1 polymorphism and risk of polycystic ovary syndrome in Turkish women. Meta Gene 2021. [DOI: 10.1016/j.mgene.2021.100959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
10
|
Kolan Ska-Dams E, Boinska J, Socha MW. Adipokine levels and carbohydrate metabolism in patients diagnosed de novo with polycystic ovary syndrome. Qatar Med J 2021; 2021:34. [PMID: 34604014 PMCID: PMC8474077 DOI: 10.5339/qmj.2021.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/07/2021] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Central obesity appears to play a major role in the pathogenesis of metabolic disorders in polycystic ovary syndrome. Insulin resistance and carbohydrate disorders are associated with dysfunctional secretion of various adipokines by the adipose tissue. OBJECTIVES This study aimed to evaluate leptin, apelin, and visfatin against a background of carbohydrate metabolism parameters in patients diagnosed de novo with polycystic ovary syndrome (PCOS). MATERIAL AND METHODS The study group consisted of 40 patients with PCOS (mean age, 29 years) diagnosed in accordance with the American Society for Reproductive Medicine criteria from 2003. The control group consisted of 37 clinically healthy women (mean age, 26 years). All controls had regular menses and no clinical or biochemical signs of hyperandrogenism. Concentrations of leptin, apelin, visfatin, and insulin were measured by immunoenzymatic methods. Glucose concentrations were determined using spectrophotometry. RESULTS Significantly higher concentrations of leptin, insulin, homeostatic model assessment for insulin resistance (HOMA-IR) index, and the immunoreactive insulin (IRI)/glucose index were found in the PCOS group than in the control group. Notably, the concentration of apelin was over five times lower in the PCOS group than in the control group. In patients with PCOS, a positive correlation was found between the concentrations of insulin and leptin and concentrations of leptin and IRI/glucose. Patients of the PCOS group with body mass index (BMI) ≥ 25 had significantly higher values of leptin, insulin, HOMA-IR index, and IRI/glucose index than patients of the PCOS group with normal BMI. In the PCOS group, a positive correlation was found between BMI and leptin concentration (r = 0.7176; p < 0.0001) and carbohydrate metabolism, such as insulin (r = 0.5524; p = 0.0003), glucose (r = 0.3843; p = 0.0157), HOMA-IR (r = 0.5895; p < 0.0001), and IRI/glucose (r = 0.3872; p = 0.0163). These findings were not observed in the control group. CONCLUSIONS (1) Increased leptin concentration observed in women diagnosed de novo with PCOS as well as positive correlations between leptin and HOMA-IR, and IRI/glucose and BMI may indicate a potential role of leptin in the reduction of tissue sensitivity to insulin. (2) Significantly lower apelin concentration in the PCOS group (>5 fold) than in the control group, associated with a concomitant increase in leptin, may also contribute to carbohydrate metabolism disorders occurring in the course of PCOS.
Collapse
Affiliation(s)
- Ewelina Kolan Ska-Dams
- Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University In Torun', Poland E-mail:
| | - Joanna Boinska
- Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University In Torun', Poland E-mail:
| | - Maciej W Socha
- Department of Obstetrics, Gynecology and Gynecological Oncology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Poland
| |
Collapse
|
11
|
Mehrabani S, Arab A, Karimi E, Nouri M, Mansourian M. Blood Circulating Levels of Adipokines in Polycystic Ovary Syndrome Patients: A Systematic Review and Meta-analysis. Reprod Sci 2021; 28:3032-3050. [PMID: 34472034 DOI: 10.1007/s43032-021-00709-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 07/30/2021] [Indexed: 12/15/2022]
Abstract
A body of studies has examined the circulating concentration of adipokines including apelin, vapin, resistin, and chemerin in polycystic ovary syndrome (PCOS) patients. However, their findings have been inconclusive. Therefore, we systematically reviewed available studies to illuminate the overall circulating concentration of adipokines in PCOS subjects. Cochrane's Library, PubMed, Scopus, and ISI Web of Science databases were searched from the earliest available date up to April 2021 for relevant articles. The quality of each study was assessed by the Newcastle-Ottawa Quality Assessment Scale. The pooled effect size was estimated based on the random effects model, and the standard mean differences (SMD) with a 95% confidence interval (CI) were reported. A total of 88 studies met the inclusion criteria and were included in the current systematic review and meta-analysis. The results of the analysis showed that serum levels of vaspin (SMD 0.69; 95% CI, 0.22 to 1.17; P = 0.004; I2 = 90.6%), chemerin (SMD 1.87; 95% CI, 1.35 to 2.40; P < 0.001; I2 = 94.4%), and resistin (SMD 0.66; 95% CI, 0.41 to 0.91; P < 0.001; I2 = 92.6%) were significantly higher in the PCOS group compared to controls. However, there was no significant difference between the PCOS and control groups in relation to apelin levels (SMD - 0.17; 95% CI, - 1.06 to 0.73; P = 0.714; I2 = 97.8%). We found that serum levels of vaspin, chemerin, and resistin were significantly higher in PCOS subjects compared with controls. It seems that these adipokines can be measured as a useful marker to predict the development of PCOS.
Collapse
Affiliation(s)
- Sanaz Mehrabani
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arman Arab
- Department of Community Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Karimi
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.,Reaserch Development Center, Arah Woman's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehran Nouri
- Department of Community Nutrition, School of Nutrition and Food Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marjan Mansourian
- Department of Biostatistics and Epidemiology, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
12
|
Raeisi T, Rezaie H, Darand M, Taheri A, Garousi N, Razi B, Roever L, Mohseni R, Hussien Mohammed S, Alizadeh S. Circulating resistin and follistatin levels in obese and non-obese women with polycystic ovary syndrome: A systematic review and meta-analysis. PLoS One 2021; 16:e0246200. [PMID: 33740002 PMCID: PMC7978365 DOI: 10.1371/journal.pone.0246200] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/15/2021] [Indexed: 12/20/2022] Open
Abstract
This meta-analysis was performed to resolve the inconsistencies regarding resistin and follistatin levels in women with polycystic ovary syndrome (PCOS) by pooling the available evidence. A systematic literature search using PubMed and Scopus was carried out through November 2020 to obtain all pertinent studies. Weighted mean differences (WMDs) with corresponding 95% confidence intervals (CIs) were calculated to evaluate the strength of the association between the levels of resistin and follistatin with PCOS in the overall and stratified analysis by obesity status. A total of 47 publications, 38 for resistin (2424 cases; 1906 controls) and 9 studies for follistatin (815 cases; 328 controls), were included in the meta-analysis. Resistin levels were significantly higher in PCOS women compared with non-PCOS controls (WMD = 1.96 ng/ml; 95%CI = 1.25-2.67, P≤0.001) as well as in obese PCOS women vs. obese controls, and in non-obese PCOS women compared with non-obese controls, but not in obese PCOS vs. non-obese PCOS patients,. A significantly increased circulating follistatin was found in PCOS patients compared with the controls (WMD = 0.44 ng/ml; 95%CI = 0.30-0.58, P≤0.001) and in non-obese PCOS women compared with non-obese controls and in obese PCOS women vs. obese controls, but, no significant difference in follistatin level was observed in obese PCOS compared with non-obese PCOS women. Significant heterogeneity and publication bias was evident for some analyses. Circulating levels of resistin and follistatin, independent of obesity status, are higher in women with PCOS compared with controls, showing that these adipokines may contribute to the pathology of PCOS.
Collapse
Affiliation(s)
- Tahereh Raeisi
- Department of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hossein Rezaie
- Department of Anatomy, School of Medicine Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Darand
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Taheri
- Department of Nutrition, Faculty of Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nazila Garousi
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahman Razi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Leonardo Roever
- Department of Clinical Research, Federal University of Uberlândia, Minas Gerais, Brazil
| | - Reza Mohseni
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Shimels Hussien Mohammed
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences-International Campus (TUMS-IC), Tehran, Iran
| | - Shahab Alizadeh
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Abstract
Polycystic ovary syndrome is a complex and heterogenous disorder involving multiple organ systems and different molecular pathways. It is tightly associated with obesity and especially abdominal obesity. As body weight reduction is the main modifiable risk factor for polycystic ovary syndrome, therapeutic approaches in overweight or obese women with polycystic ovary syndrome have been developed. Liraglutide is a glucagon-like peptide-1 receptor agonist that promotes sustained weight loss, as well as abdominal fat reduction, in individuals with obesity, prediabetes, and type 2 diabetes mellitus. The majority of current clinical studies have demonstrated that liraglutide therapy achieved significant reductions in body weight, body mass index, and abdominal circumference in overweight and obese women with polycystic ovary syndrome. Liraglutide therapy promoted significant improvements in free testosterone and sex hormone-binding globulin levels in some studies. Important metabolic and hormonal improvements were also reported after the combination of liraglutide with metformin. Increased menstrual frequency, as well as potential positive effects in reproduction, were described. However, the small number of participants, short duration, and low daily liraglutide dose are some of the main limitations of these studies. Larger and longer, multi-centred, double-blind, placebo-controlled trials of liraglutide monotherapy or combination therapy, with prolonged post-interventional monitoring, are crucially anticipated. Metabolic, hormonal, and reproductive primary outcomes should be uniformly addressed, to tailor future targeted treatment approaches, according to the patient phenotype and needs. This will improve long-term therapeutic outcomes in this population.
Collapse
|
14
|
Nasiadek M, Stragierowicz J, Klimczak M, Kilanowicz A. The Role of Zinc in Selected Female Reproductive System Disorders. Nutrients 2020; 12:E2464. [PMID: 32824334 PMCID: PMC7468694 DOI: 10.3390/nu12082464] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022] Open
Abstract
Zinc is an essential microelement that plays many important functions in the body. It is crucial for the regulation of cell growth, hormone release, immunological response and reproduction. This review focuses on its importance in the reproductive system of women of reproductive and postmenopausal ages, not including its well described role in pregnancy. Only recently, attention has been drawn to the potential role of zinc in polycystic ovary syndrome (PCOS), dysmenorrhea, or endometriosis. This review is mainly based on 36 randomized, controlled studies on reproductive, pre- and post-menopausal populations of women and on research trying to explain the potential impact of zinc and its supplementation in the etiology of selected female reproductive system disorders. In women with PCOS, zinc supplementation has a positive effect on many parameters, especially those related to insulin resistance and lipid balance. In primary dysmenorrhea, zinc supplementation before and during each menstrual cycle seems to be an important factor reducing the intensity of menstrual pain. On the other hand, little is known of the role of zinc in endometriosis and in postmenopausal women. Therefore, further studies explaining the potential impact of zinc and its supplementation on female reproductive system would be highly advisable and valuable.
Collapse
Affiliation(s)
- Marzenna Nasiadek
- Department of Toxicology, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland; (J.S.); (M.K.)
| | | | | | - Anna Kilanowicz
- Department of Toxicology, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland; (J.S.); (M.K.)
| |
Collapse
|
15
|
Stepto NK, Hiam D, Gibson-Helm M, Cassar S, Harrison CL, Hutchison SK, Joham AE, Canny BJ, Moreno-Asso A, Strauss BJ, Hatzirodos N, Rodgers RJ, Teede HJ. Exercise and insulin resistance in PCOS: muscle insulin signalling and fibrosis. Endocr Connect 2020; 9:346-359. [PMID: 32229703 PMCID: PMC7219141 DOI: 10.1530/ec-19-0551] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 03/31/2020] [Indexed: 12/05/2022]
Abstract
OBJECTIVE Mechanisms of insulin resistance in polycystic ovary syndrome (PCOS) remain ill defined, contributing to sub-optimal therapies. Recognising skeletal muscle plays a key role in glucose homeostasis we investigated early insulin signalling, its association with aberrant transforming growth factor β (TGFβ)-regulated tissue fibrosis. We also explored the impact of aerobic exercise on these molecular pathways. METHODS A secondary analysis from a cross-sectional study was undertaken in women with (n = 30) or without (n = 29) PCOS across lean and overweight BMIs. A subset of participants with (n = 8) or without (n = 8) PCOS who were overweight completed 12 weeks of aerobic exercise training. Muscle was sampled before and 30 min into a euglycaemic-hyperinsulinaemic clamp pre and post training. RESULTS We found reduced signalling in PCOS of mechanistic target of rapamycin (mTOR). Exercise training augmented but did not completely rescue this signalling defect in women with PCOS. Genes in the TGFβ signalling network were upregulated in skeletal muscle in the overweight women with PCOS but were unresponsive to exercise training except for genes encoding LOX, collagen 1 and 3. CONCLUSIONS We provide new insights into defects in early insulin signalling, tissue fibrosis, and hyperandrogenism in PCOS-specific insulin resistance in lean and overweight women. PCOS-specific insulin signalling defects were isolated to mTOR, while gene expression implicated TGFβ ligand regulating a fibrosis in the PCOS-obesity synergy in insulin resistance and altered responses to exercise. Interestingly, there was little evidence for hyperandrogenism as a mechanism for insulin resistance.
Collapse
Affiliation(s)
- N K Stepto
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
- Monash Centre for Health Research and Implementation, Monash University, Clayton, Victoria, Australia
- Australian Institute for Musculoskeletal Science, Victoria University, Melbourne, Victoria, Australia
- Medicine-Western Health, Faculty of Medicine, Dentistry and Health Science, Melbourne University, Melbourne, Victoria, Australia
| | - D Hiam
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - M Gibson-Helm
- Monash Centre for Health Research and Implementation, Monash University, Clayton, Victoria, Australia
| | - S Cassar
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - C L Harrison
- Monash Centre for Health Research and Implementation, Monash University, Clayton, Victoria, Australia
| | - S K Hutchison
- Monash Centre for Health Research and Implementation, Monash University, Clayton, Victoria, Australia
| | - A E Joham
- Monash Centre for Health Research and Implementation, Monash University, Clayton, Victoria, Australia
| | - B J Canny
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - A Moreno-Asso
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science, Victoria University, Melbourne, Victoria, Australia
| | - B J Strauss
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
- Division of Diabetes, Endocrinology & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - N Hatzirodos
- The Robinson Research Institute, School of Medicine, The University of Adelaide, Adelaide, Australia
| | - R J Rodgers
- The Robinson Research Institute, School of Medicine, The University of Adelaide, Adelaide, Australia
| | - H J Teede
- Monash Centre for Health Research and Implementation, Monash University, Clayton, Victoria, Australia
- Diabetes and Endocrine Units, Monash Health, Clayton, Victoria, Australia
| |
Collapse
|
16
|
Moffett RC, Naughton V. Emerging role of GIP and related gut hormones in fertility and PCOS. Peptides 2020; 125:170233. [PMID: 31935429 DOI: 10.1016/j.peptides.2019.170233] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023]
Abstract
Gastric inhibitory polypeptide (GIP) is best known as an incretin hormone released by enteroendocrine K-cells in response to feeding and stimulates insulin release to regulate blood glucose and nutrient homeostasis. More recently GIP has been ascribed a positive role in lipid metabolism, bone strength, cardiovascular function and cognition. The present paper considers an emerging role of GIP and related gut hormones in fertility and especially polycystic ovarian syndrome (PCOS). Key evidence concerns restoration of fertility in women with gross obesity and PCOS following bariatric surgery. This is considered to reflect indirect effects mediated by alleviation of insulin resistance together with possible direct effects of surgically induced changes of GIP, GLP-1 and related peptide hormones on ovaries and the hypothalamic-pituitary-adrenal axis. Further studies are required to determine inter-relationships between the hormones and cellular mechanisms involved but these observations suggest that GIP and other gut may provide a novel therapeutic approach for PCOS and other reproductive disorders.
Collapse
Affiliation(s)
- R Charlotte Moffett
- SAAD Centre for Pharmacy and Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, Northern Ireland, UK.
| | - Violetta Naughton
- SAAD Centre for Pharmacy and Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, Northern Ireland, UK
| |
Collapse
|
17
|
Ilie IR. Neurotransmitter, neuropeptide and gut peptide profile in PCOS-pathways contributing to the pathophysiology, food intake and psychiatric manifestations of PCOS. Adv Clin Chem 2019; 96:85-135. [PMID: 32362321 DOI: 10.1016/bs.acc.2019.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a major health problem with a heterogeneous hormone-imbalance and clinical presentation across the lifespan of women. Increased androgen production and abnormal gonadotropin-releasing hormone (GnRH) release and gonadotropin secretion, resulting in chronic anovulation are well-known features of the PCOS. The brain is both at the top of the neuroendocrine axis regulating ovarian function and a sensitive target of peripheral gonadal hormones and peptides. Current literature illustrates that neurotransmitters regulate various functions of the body, including reproduction, mood and body weight. Neurotransmitter alteration could be one of the reasons for disturbed GnRH release, consequently directing the ovarian dysfunction in PCOS, since there is plenty evidence for altered catecholamine metabolism and brain serotonin or opioid activity described in PCOS. Further, the dysregulated neurotransmitter and neuropeptide profile in PCOS could also be the reason for low self-esteem, anxiety, mood swings and depression or obesity, features closely associated with PCOS women. Can these altered central brain circuits, or the disrupted gut-brain axis be the tie that would both explain and link the pathogenesis of this disorder, the occurrence of depression, anxiety and other mood disorders as well as of obesity, insulin resistance and abnormal appetite in PCOS? This review intends to provide the reader with a comprehensive overview of what is known about the relatively understudied, but very complex role that neurotransmitters, neuropeptides and gut peptides play in PCOS. The answer to the above question may help the development of drugs to specifically target these central and peripheral circuits, thereby providing a valuable treatment for PCOS patients that present to the clinic with GnRH/LH hypersecretion, obesity or psychiatric manifestations.
Collapse
Affiliation(s)
- Ioana R Ilie
- Department of Endocrinology, University of Medicine and Pharmacy 'Iuliu-Hatieganu', Cluj-Napoca, Romania.
| |
Collapse
|
18
|
Ali AT, Guidozzi F. Midlife women's health consequences associated with polycystic ovary syndrome. Climacteric 2019; 23:116-122. [PMID: 31657237 DOI: 10.1080/13697137.2019.1679111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common female endocrinopathies. Its symptoms may appear as early as adolescence and may include irregular menstrual periods, amenorrhea, hirsutism and obesity. Regardless of their phenotypic appearance, women with PCOS are metabolically obese. PCOS is associated with metabolic syndrome, type 2 diabetes, depression, cardiovascular disease and gynecological cancers. The metabolic disorders in obese women with PCOS are invariably due to insulin resistance, while inflammation, oxidative stress and possible interaction with environmental factors are among the features linking women with PCOS alone to metabolic disorders. The current review aims to highlight the relationship between PCOS and midlife women's health complications.
Collapse
Affiliation(s)
- A T Ali
- Department of Chemical Pathology, NHLS, Tygerberg Hospital and Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - F Guidozzi
- Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
19
|
Metabolic Cytokines at Fasting and During Macronutrient Challenges: Influence of Obesity, Female Androgen Excess and Sex. Nutrients 2019; 11:nu11112566. [PMID: 31652917 PMCID: PMC6893420 DOI: 10.3390/nu11112566] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/11/2019] [Accepted: 09/15/2019] [Indexed: 12/17/2022] Open
Abstract
Scope: Cytokines have pleiotropic functions within the organism and their levels may be influenced by obesity, visceral adiposity and sex hormones. Diet composition may also affect their systemic concentrations during fasting and in the postprandial period. Hence, we studied the influence of sex steroids and obesity on the circulating levels of a panel of metabolic cytokines in the fasting state and after single macronutrient challenges. Methods: On alternate days we submitted 17 women with polycystic ovary syndrome (PCOS) (9 non-obese, 8 obese), 17 non-hyperandrogenic control women (9 non-obese, 8 obese) and 19 control men (10 non-obese, 9 obese) to isocaloric oral glucose, lipid and protein loads. Serum levels of omentin-1, vaspin, lipocalin-2, adipsin, PAI-1, chemerin, FGF-21 and FGF-23 were determined by Luminex multiplex technology. Results: During fasting, obese patients presented higher levels of PAI-1, chemerin and adipsin but decreased FGF-23 and omentin-1 compared with non-obese subjects. Vaspin showed sexual dimorphism with lower levels in men than women with PCOS and female controls. Following macronutrient ingestion, most metabolic cytokines presented a similar physiological response consisting of a decrease in circulating concentrations, which was inversely associated with the fasting levels of these molecules. Protein intake caused the major postprandial decrease whereas glucose did not significantly reduce PAI-1, FGF-23 and vaspin, and even increased FGF-21. Regardless of the macronutrient administered, vaspin levels showed a larger reduction in non-obese individuals while the decrease in PAI-1 was particularly noticeable in the obese subgroup. The postprandial reductions of omentin-1 and FGF-23 after glucose and protein loads were influenced by obesity. No major differences were found between patients with PCOS and male and female controls. Conclusions: Obesity, but not PCOS or sex, markedly influences metabolic cytokine levels at fasting and after macronutrient ingestion. The observed postprandial decrease in their circulating concentrations might represent a physiological compensatory mechanism against food-induced inflammation and oxidative stress. This mechanism is altered by obesity and is differently modulated by macronutrients, suggesting a larger contribution of glucose to stressful postprandial responses.
Collapse
|
20
|
Hu Y, Wang J, Zhou Y, Xie H, Yan X, Chu X, Chen W, Liu Y, Wang X, Wang J, Zhang A, Han S. Peptidomics analysis of umbilical cord blood reveals potential preclinical biomarkers for neonatal respiratory distress syndrome. Life Sci 2019; 236:116737. [PMID: 31505194 DOI: 10.1016/j.lfs.2019.116737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 02/04/2023]
Abstract
AIMS The purpose of this study was to investigate the pathophysiology and discover novel predictors of neonatal respiratory distress syndrome (NRDS) from a peptidomics perspective. MAIN METHODS Comparative profiling of umbilical cord blood from NRDS and control patients was performed by liquid chromatography tandem mass spectrometry technology. The underlying biological functions of the differentially expressed peptides (DEPs) were predicted by Gene Ontology (GO) and KEGG pathway analyses. The interactions of DEPs and their precursor proteins were explored by ingenuity pathway analysis (IPA). The sources and stability of DEPs were determined by online databases, including UniProt, SMART and ProtParam tool. KEY FINDINGS A total of 251 DEPs were identified, of which 139 peptides were upregulated, and 112 peptides were downregulated (fold change ≥2.0, P < 0.05). These DEPs were predicted to be associated with respiratory failure, atelectasis, and morphogenesis of endothelial cells. These processes indicated that DEPs may play a role in NRDS. Among them, eleven stable DEPs might be used as preclinical biomarkers. SIGNIFICANCE Our findings improve our understanding of NRDS and facilitate the discovery of candidate diagnostic biomarkers for NRDS from the perspective of peptidomics.
Collapse
Affiliation(s)
- Yin Hu
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Juan Wang
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Yahui Zhou
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Hanying Xie
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China; The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Xiangyun Yan
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Xue Chu
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Wenjuan Chen
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Yiwen Liu
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China; The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Xingyun Wang
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Jun Wang
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China; The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China.
| | - Aiqing Zhang
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China; Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, 262 Zhongshan North Road, Nanjing, Jiangsu 210003, China.
| | - Shuping Han
- Department of Pediatrics, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China.
| |
Collapse
|
21
|
Ferjan S, Jensterle M, Oblak T, Zitnik IP, Marc J, Goricar K, Dolzan V, Janez A. An impaired glucagon-like peptide-1 response is associated with prediabetes in polycystic ovary syndrome with obesity. J Int Med Res 2019; 47:4691-4700. [PMID: 31438749 PMCID: PMC6833370 DOI: 10.1177/0300060519865351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Objective Impaired glucose homeostasis in polycystic ovary syndrome (PCOS) is associated with obesity, age, and disease phenotype. This study aimed to investigate the glucagon-like peptide-1 (GLP-1) response in patients with obesity and PCOS with normal glucose tolerance (NGT) or prediabetes. Methods Twenty-six women with obesity and PCOS were included. Thirteen women had NGT and 13 had prediabetes. Serum glucose, insulin, and GLP-1 levels were measured during an oral glucose tolerance test. Beta-cell function and insulin resistance were determined. Results Women with prediabetes had significantly lower GLP-1 levels than did those with NGT after a glucose load. GLP-1 levels <3.02 pM at 120 minutes were associated with prediabetes. Women with prediabetes had a lower oral glucose insulin sensitivity (OGIS) index and greater amount of visceral adipose tissue than did those with NGT. Plasma GLP-1 levels at 120 minutes were correlated with visceral adiposity and the OGIS index. A change in GLP-1 levels was correlated with a family history of type 2 diabetes. Conclusion The GLP-1 response is lower in patients with obesity, PCOS, and prediabetes than in those with obesity, PCOS, and NGT. Further investigation of the GLP-1 response as a potential separate risk factor for prediabetes in PCOS is required.
Collapse
Affiliation(s)
- Simona Ferjan
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre, SI-1000 Ljubljana, Slovenia.,University of Ljubljana Faculty of Medicine, SI-1000 Ljubljana, Slovenia
| | - Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre, SI-1000 Ljubljana, Slovenia.,University of Ljubljana Faculty of Medicine, SI-1000 Ljubljana, Slovenia
| | - Tjasa Oblak
- University of Ljubljana Faculty of Medicine, SI-1000 Ljubljana, Slovenia
| | | | - Janja Marc
- University of Ljubljana Faculty of Pharmacy, SI-1000 Ljubljana, Slovenia
| | - Katja Goricar
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Pharmacogenetics Laboratory, SI-1000 Ljubljana, Slovenia
| | - Vita Dolzan
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Pharmacogenetics Laboratory, SI-1000 Ljubljana, Slovenia
| | - Andrej Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre, SI-1000 Ljubljana, Slovenia.,University of Ljubljana Faculty of Medicine, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
22
|
Baldani DP, Skrgatic L, Kasum M, Zlopasa G, Kralik Oguic S, Herman M. Altered leptin, adiponectin, resistin and ghrelin secretion may represent an intrinsic polycystic ovary syndrome abnormality. Gynecol Endocrinol 2019; 35:401-405. [PMID: 30623695 DOI: 10.1080/09513590.2018.1534096] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The aim of the study was to investigate whether altered adipose tissue secretion of various adipokines is secondary to obesity, hyperandrogenism, and hyperinsulinemia or intrinsic to polycystic ovary syndrome (PCOS). This cross-sectional study included 151 women diagnosed with PCOS by the Rotterdam criteria and 95 healthy women matched by age, body mass index (BMI), and waist-to-hip ratio (WHR). Clinical, biochemical, and hormonal characteristics were assessed. Serum concentrations of ghrelin and adiponectin were found to be significantly lower and concentrations of leptin and resistin significantly higher in women with PCOS than in healthy women matched by age, BMI, and WHR. A PCOS diagnosis made the largest contribution to predicting serum levels of leptin, adiponectin, resistin, and ghrelin in all stepwise multiple regression models, which included PCOS diagnosis, BMI, WHR, luteinizing hormone, total testosterone, free testosterone and homeostatic model assessment of insulin resistance as independent predictors. Leptin, adiponectin, ghrelin and resistin levels may serve as independent biomarkers for the diagnosis of PCOS.
Collapse
Affiliation(s)
- Dinka Pavičić Baldani
- a School of Medicine, Department of Obstetrics and Gynecology, Division of Gynecological Endocrinology and Reproductive Medicine, Clinical Hospital Centre Zagreb , University of Zagreb , Zagreb , Croatia
| | - Lana Skrgatic
- a School of Medicine, Department of Obstetrics and Gynecology, Division of Gynecological Endocrinology and Reproductive Medicine, Clinical Hospital Centre Zagreb , University of Zagreb , Zagreb , Croatia
| | - Miro Kasum
- a School of Medicine, Department of Obstetrics and Gynecology, Division of Gynecological Endocrinology and Reproductive Medicine, Clinical Hospital Centre Zagreb , University of Zagreb , Zagreb , Croatia
| | - Gordan Zlopasa
- a School of Medicine, Department of Obstetrics and Gynecology, Division of Gynecological Endocrinology and Reproductive Medicine, Clinical Hospital Centre Zagreb , University of Zagreb , Zagreb , Croatia
| | - Saša Kralik Oguic
- b Clinical Institute of Laboratory Diagnostics , Clinical Hospital Centre , Zagreb , Croatia
| | - Mislav Herman
- a School of Medicine, Department of Obstetrics and Gynecology, Division of Gynecological Endocrinology and Reproductive Medicine, Clinical Hospital Centre Zagreb , University of Zagreb , Zagreb , Croatia
| |
Collapse
|
23
|
Bastard JP, Couffignal C, Fellahi S, Bard JM, Mentre F, Salmon D, Katlama C, Raffi F, Leport C, Capeau J. Diabetes and dyslipidaemia are associated with oxidative stress independently of inflammation in long-term antiretroviral-treated HIV-infected patients. DIABETES & METABOLISM 2019; 45:573-581. [PMID: 30862472 DOI: 10.1016/j.diabet.2019.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 02/15/2019] [Accepted: 02/24/2019] [Indexed: 12/31/2022]
Abstract
AIM Ageing HIV-infected patients controlled by antiretroviral therapy (ART) frequently present age-related comorbidities, such as cardiovascular (CV) events, diabetes, dyslipidaemia, hypertension and chronic kidney disease (CKD). The prevalence of these comorbidities was evaluated in a cohort of long-term-monitored ART-controlled HIV-infected patients, then followed by a search into whether oxidative stress, like inflammation, might be associated with metabolic parameters and/or comorbidities. METHODS Included were 352 long-term ART patients who started with protease inhibitors (PIs) in 1997-1999. They were evaluated at their final visit, 11 years later, for previous CV events, prevalence of diabetes, LDL-related and atherogenic (high TG/HDL) dyslipidaemias, hypertension and CKD. Also measured were circulating biomarkers to explore oxidative stress (Lp-PLA2, oxLDL, oxLDL/LDL ratio, paraoxonase and arylesterase activities), inflammation/immune activation (hsCRP, hsIL-6, D dimer, soluble CD14, β2 microglobulin, cystatin C), adipokines and insulin resistance. Levels were compared in patients with and without each comorbidity or condition using non-parametric correlation tests and multivariate adjusted analyses. RESULTS At the final visit, 81.5% of patients were male and were aged (median, IQR) 49 years (45-56); BMI was 23.0 kg/m2 (21.1-25.4), CD4+ lymphocytes were 620 cells/mm3 (453-790) and 91.5% had undetectable HIV-1 viral loads. The prevalence of diabetes was 11%, and LDL-related dyslipidaemia 28%, atherogenic dyslipidaemia 9%, hypertension 28%, CKD 9% and previous CV events 9%. Diabetes and atherogenic dyslipidaemia were associated with increased oxidative stress and independently with inflammation. LDL-related dyslipidaemia and impaired fasting glucose were associated with increased oxidative stress. No association of these biomarkers was detected with hypertension, CKD and previous CV events. CONCLUSION In long-term-treated HIV-infected patients with frequent comorbid conditions, oxidative stress could be contributing to diabetes and LDL-related and atherogenic dyslipidaemias independently of inflammation.
Collapse
Affiliation(s)
- J-P Bastard
- Faculty of medicine, Sorbonne université, inserm UMR_S938, ICAN, AP-HP, hôpital Tenon, 27, rue Chaligny, 75571 Paris cedex 12, Paris, France
| | - C Couffignal
- Université Paris Diderot, Sorbonne Paris Cité, inserm UMR_S1137, COREB APHP, 16, rue Henri-Huchard, 75890 Paris cedex 18, France
| | - S Fellahi
- Faculty of medicine, Sorbonne université, inserm UMR_S938, ICAN, AP-HP, hôpital Tenon, 27, rue Chaligny, 75571 Paris cedex 12, Paris, France
| | - J-M Bard
- UFR des sciences pharmaceutiques et biologiques, MMS - EA 2160, IUML FR3473 CNRS, Nantes and institut de cancérologie de l'Ouest, 4, rue Bras France, BP61112, 44035 Nantes cedex 1, France
| | - F Mentre
- Université Paris Diderot, Sorbonne Paris Cité, inserm UMR_S1137, COREB APHP, 16, rue Henri-Huchard, 75890 Paris cedex 18, France
| | - D Salmon
- Service des maladies infectieuses et tropicales, hôpital Cochin, AP-HP, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - C Katlama
- Service de maladies infectieuses et tropicales hôpital Pitié-Salpêtrière, AP-HP, Sorbonne Universités, UPMC Université Paris-6, inserm UMR_S1136 IPLESP, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - F Raffi
- Service des maladies infectieuses et tropicales, inserm CIC 1413, CHU de Nantes, 1, place Alexis-Ricordeau, 44000 Nantes, France
| | - C Leport
- Université Paris Diderot, Sorbonne Paris Cité, inserm UMR_S1137, COREB APHP, 16, rue Henri-Huchard, 75890 Paris cedex 18, France
| | - J Capeau
- Faculty of medicine, Sorbonne université, inserm UMR_S938, ICAN, AP-HP, hôpital Tenon, 27, rue Chaligny, 75571 Paris cedex 12, Paris, France.
| | | |
Collapse
|
24
|
Esmaeilinezhad Z, Babajafari S, Sohrabi Z, Eskandari MH, Amooee S, Barati-Boldaji R. Effect of synbiotic pomegranate juice on glycemic, sex hormone profile and anthropometric indices in PCOS: A randomized, triple blind, controlled trial. Nutr Metab Cardiovasc Dis 2019; 29:201-208. [PMID: 30538082 DOI: 10.1016/j.numecd.2018.07.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Polycystic Ovarian Syndrome is a common reproductive, endocrine, and metabolic disease in women. Pomegranate juice, known as a rich source of phytochemicals with high antioxidant activity, enriched with probiotic may improve PCOS. METHODS AND RESULTS A randomized, controlled, triple-blinded, parallel trial study was performed in PCOS patients (n = 92). Three treatment groups (23 patients each) received 2 L of synbiotic pomegranate juice (SPJ), pomegranate juice (PJ), and synbiotic beverage (SB) weekly. The control group (23 patients) received 2 L of placebo beverage weekly. Primary outcome was any change in insulin resistance and secondary outcomes were fasting blood sugar (FBS), insulin sensitivity, testosterone, luteinizing hormone (LH), follicle stimulating hormone (FSH), body mass index (BMI), waist and hip circumference, from baseline to the end of the trial. At the end of the study, 86 patients were analyzed. There was significant change in insulin resistance in the SPJ and SB groups. Insulin sensitivity increased significantly in the SPJ and SB groups. Insulin also changed significantly in the SPJ and SB groups. BMI, weight and waist circumference decreased significantly in the SPJ and SB groups. Testosterone level also decreased significantly in the SPJ and SB groups. There was no significant change in FPG, LH and FSH in any of the groups. CONCLUSION SPJ in the form of a new beverage can improve insulin resistance, insulin, testosterone level, BMI, weight and waist circumference in PCOS. This trial was registered in Iranian Registry of Clinical Trials, with number: 25272.
Collapse
Affiliation(s)
- Z Esmaeilinezhad
- Nutrition Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - S Babajafari
- Nutrition Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Z Sohrabi
- Nutrition Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - M-H Eskandari
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - S Amooee
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - R Barati-Boldaji
- Nutrition Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
25
|
Zadeh Modarres S, Heidar Z, Foroozanfard F, Rahmati Z, Aghadavod E, Asemi Z. The Effects of Selenium Supplementation on Gene Expression Related to Insulin and Lipid in Infertile Polycystic Ovary Syndrome Women Candidate for In Vitro Fertilization: a Randomized, Double-Blind, Placebo-Controlled Trial. Biol Trace Elem Res 2018; 183:218-225. [PMID: 28875327 DOI: 10.1007/s12011-017-1148-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 08/30/2017] [Indexed: 12/12/2022]
Abstract
UNLABELLED This study was conducted to evaluate the effects of selenium supplementation on gene expression related to insulin and lipid in infertile women with polycystic ovary syndrome (PCOS) candidate for in vitro fertilization (IVF). This randomized double-blind, placebo-controlled trial was conducted among 40 infertile women with PCOS candidate for IVF. Subjects were randomly allocated into two groups to intake either 200-μg selenium (n = 20) or placebo (n = 20) per day for 8 weeks. Gene expression levels related to insulin and lipid were quantified in lymphocytes of women with PCOS candidate for IVF with RT-PCR method. Results of RT-PCR demonstrated that after the 8-week intervention, compared with the placebo, selenium supplementation upregulated gene expression of peroxisome proliferator-activated receptor gamma (PPAR-γ) (1.06 ± 0.15-fold increase vs. 0.94 ± 0.18-fold reduction, P = 0.02) and glucose transporter 1 (GLUT-1) (1.07 ± 0.20-fold increase vs. 0.87 ± 0.18-fold reduction, P = 0.003) in lymphocytes of women with PCOS candidate for IVF. In addition, compared with the placebo, selenium supplementation downregulated gene expression of low-density lipoprotein receptor (LDLR) (0.88 ± 0.17-fold reduction vs. 1.05 ± 0.22-fold increase, P = 0.01) in lymphocytes of women with PCOS candidate for IVF. We did not observe any significant effect of selenium supplementation on gene expression levels of lipoprotein(a) [LP(a)] in lymphocytes of women with PCOS candidate for IVF. Overall, selenium supplementation for 8 weeks in lymphocytes of women with infertile PCOS candidate for IVF significantly increased gene expression levels of PPAR-γ and GLUT-1 and significantly decreased gene expression levels of LDLR, but did not affect LP(a). CLINICAL TRIAL REGISTRATION NUMBER http://www.irct.ir : IRCT201704245623N113.
Collapse
Affiliation(s)
- Shahrzad Zadeh Modarres
- Infertility and Reproductive Health Research Center (IRHRC), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Heidar
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Foroozanfard
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Rahmati
- Infertility and Reproductive Health Research Center (IRHRC), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
26
|
Chromium supplementation does not improve weight loss or metabolic and hormonal variables in patients with polycystic ovary syndrome: A systematic review. Nutr Res 2018; 56:1-10. [PMID: 30055769 DOI: 10.1016/j.nutres.2018.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 04/03/2018] [Accepted: 04/08/2018] [Indexed: 12/14/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrinopathy in women of reproductive age, and recently, chromium was discussed as an adjuvant way to manage it. Herein, a systematic review was conducted which centered on the effects of chromium on ovarian physiology with a focus on body mass index, as well as hormonal and metabolic dysfunctions in women suffering from PCOS. This review was performed using the guidelines from Preferred Reporting Items for Systematic Reviews. Clinical trials investigating chromium in women with PCOS with outcome measures related to metabolic and hormonal status were included. The search was conducted using PubMed, Scopus, and Google Scholar databases for clinical trials in the English language from the inception of the resources until May 2017 with the terms: chromium, chromium picolinate, CrP, polycystic ovary syndrome, PCOS, and sclerocystic ovary syndrome. The search resulted in 89 articles, and after inclusion and exclusion criteria were applied, 6 articles were selected for analysis. Two studies that evaluated the effect of chromium on body weight or body mass index reported no effect. Another study reported the beneficial effect of chromium on weight reduction. It seems that the effect of chromium in the reduction of blood glucose is insignificant, and results are inconsistent in relation to dyslipidemia. With regard to the effects of chromium on concentrations of sex hormones, a longer duration of treatment is needed to produce significant changes. The articles reviewed demonstrated that chromium supplementation has limited effects on weight reduction, glucose control, lipid profile, and hormonal disturbance of women with PCOS; however, more studies are needed due to the clinical changes observed in patients with PCOS after chromium supplementation.
Collapse
|
27
|
Kumarendran B, O’Reilly MW, Manolopoulos KN, Toulis KA, Gokhale KM, Sitch AJ, Wijeyaratne CN, Coomarasamy A, Arlt W, Nirantharakumar K. Polycystic ovary syndrome, androgen excess, and the risk of nonalcoholic fatty liver disease in women: A longitudinal study based on a United Kingdom primary care database. PLoS Med 2018; 15:e1002542. [PMID: 29590099 PMCID: PMC5873722 DOI: 10.1371/journal.pmed.1002542] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 02/21/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Androgen excess is a defining feature of polycystic ovary syndrome (PCOS), which affects 10% of women and represents a lifelong metabolic disorder, with increased risk of type 2 diabetes, hypertension, and cardiovascular events. Previous studies have suggested an increased risk of nonalcoholic fatty liver disease (NAFLD) in individuals with PCOS and implicated androgen excess as a potential driver. METHODS AND FINDINGS We carried out a retrospective longitudinal cohort study utilizing a large primary care database in the United Kingdom, evaluating NAFLD rates in 63,120 women with PCOS and 121,064 age-, body mass index (BMI)-, and location-matched control women registered from January 2000 to May 2016. In 2 independent cohorts, we also determined the rate of NAFLD in women with a measurement of serum testosterone (n = 71,061) and sex hormone-binding globulin (SHBG; n = 49,625). We used multivariate Cox models to estimate the hazard ratio (HR) for NAFLD and found that women with PCOS had an increased rate of NAFLD (HR = 2.23, 95% CI 1.86-2.66, p < 0.001), also after adjusting for BMI or dysglycemia. Serum testosterone >3.0 nmol/L was associated with an increase in NAFLD (HR = 2.30, 95% CI 1.16-4.53, p = 0.017 for 3-3.49 nmol/L and HR = 2.40, 95% CI 1.24-4.66, p = 0.009 for >3.5 nmol/L). Mirroring this finding, SHBG <30 nmol/L was associated with increased NAFLD hazard (HR = 4.75, 95% CI 2.44-9.25, p < 0.001 for 20-29.99 nmol/L and HR = 4.98, 95% CI 2.45-10.11, p < 0.001 for <20 nmol/L). Limitations of this study include its retrospective nature, absence of detailed information on criteria used to diagnosis PCOS and NAFLD, and absence of data on laboratory assays used to measure serum androgens. CONCLUSIONS We found that women with PCOS have an increased rate of NAFLD. In addition to increased BMI and dysglycemia, androgen excess contributes to the development of NAFLD in women with PCOS. In women with PCOS-related androgen excess, systematic NAFLD screening should be considered.
Collapse
Affiliation(s)
- Balachandran Kumarendran
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
- Department of Public Health, Faculty of Medicine, University of Kelaniya, Kelaniya, Sri Lanka
| | - Michael W. O’Reilly
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Konstantinos N. Manolopoulos
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Konstantinos A. Toulis
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Krishna M. Gokhale
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Alice J. Sitch
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Chandrika N. Wijeyaratne
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Arri Coomarasamy
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
- * E-mail: (WA); (KN)
| | - Krishnarajah Nirantharakumar
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
- * E-mail: (WA); (KN)
| |
Collapse
|
28
|
Shorakae S, Abell SK, Hiam DS, Lambert EA, Eikelis N, Jona E, Sari CI, Stepto NK, Lambert GW, de Courten B, Teede HJ. High-molecular-weight adiponectin is inversely associated with sympathetic activity in polycystic ovary syndrome. Fertil Steril 2018; 109:532-539. [DOI: 10.1016/j.fertnstert.2017.11.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 01/22/2023]
|
29
|
de Medeiros SF, Angelo LCA, de Medeiros MAS, Banhara CR, Barbosa BB, Yamamoto MMW. The Role of C-Peptide as Marker of Cardiometabolic Risk in Women With Polycystic Ovary Syndrome: A Controlled Study. J Clin Med Res 2018; 10:260-267. [PMID: 29416587 PMCID: PMC5798275 DOI: 10.14740/jocmr3325w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 01/12/2018] [Indexed: 12/20/2022] Open
Abstract
Background The aim of this study was to examine the role of C-peptide as a biological marker of cardiometabolic risk in polycystic ovary syndrome (PCOS). Methods This case-control study enrolled 385 PCOS patients and 240 normal cycling women. Anthropometric and clinical variables were taken at first visit. Fasting C-peptide, glucose, lipids, and hormone measurements were performed. Simple and multiple correlations between C-peptide and other variables associated with dysmetabolism and cardiovascular disease were examined. Results C-peptide was well correlated with several anthropometric, metabolic, and endocrine parameters. In PCOS patients, stepwise multiple regression including C-peptide as the criterion variable and other predictors of cardiovascular disease risk provided a significant model in which the fasting C-peptide/glucose ratio, glucose, body weight, and free estrogen index (FEI) were retained (adjusted R2 = 0.988, F = 7.161, P = 0.008). Conclusion C-peptide levels alone or combined with C-peptide/glucose ratio, glucose, body weight, and FEI provided a significant model to identify PCOS patients with higher risk of future cardiometabolic diseases.
Collapse
Affiliation(s)
- Sebastiao Freitas de Medeiros
- Department of Gynecology and Obstetrics, Medical School, Federal University of Mato Grosso, Cuiaba, MT, Brazil.,Tropical Institute of Reproductive Medicine, Cuiaba, MT, Brazil
| | - Laura Camila Antunes Angelo
- Department of Gynecology and Obstetrics, Medical School, Federal University of Mato Grosso, Cuiaba, MT, Brazil
| | | | | | | | | |
Collapse
|
30
|
Mirmasoumi G, Fazilati M, Foroozanfard F, Vahedpoor Z, Mahmoodi S, Taghizadeh M, Esfeh N, Mohseni M, Karbassizadeh H, Asemi Z. The Effects of Flaxseed Oil Omega-3 Fatty Acids Supplementation on Metabolic Status of Patients with Polycystic Ovary Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Exp Clin Endocrinol Diabetes 2017; 126:222-228. [DOI: 10.1055/s-0043-119751] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Abstract
Objective This study was conducted to evaluate the effects of flaxseed oil omega-3 fatty acids supplementation on metabolic status of patients with polycystic ovary syndrome (PCOS).
Methods This randomized double-blind, placebo-controlled trial was conducted on 60 women with PCOS according to the Rotterdam criteria aged 18–40 years old. Participants were randomly assigned into two groups to receive either 1,000 mg flaxseed oil omega-3 fatty acids (n=30) or placebo (n=30) twice a day for 12 weeks. Metabolic, endocrine, inflammatory factors were quantified at baseline and after the 12-week intervention.
Results After the 12-week intervention, compared to the placebo, flaxseed oil omega-3 supplementation significantly decreased insulin values (−2.6±7.7 vs.+1.3±3.9 µIU/mL, P=0.01), homeostasis model of assessment-estimated insulin resistance (−0.7±1.7 vs.+0.3±0.9, P=0.01), mF-G scores (−1.2±1.7 vs. -0.1±0.4, P=0.001), and increased quantitative insulin sensitivity check index (+0.01±0.02 vs. −0.01±0.02, P=0.01). In addition, supplementation with flaxseed oil omega-3 resulted in significant decreases in serum triglycerides (−5.1±20.9 vs.+9.7±26.1 mg/dL, P=0.01), VLDL-cholesterol (−1.0±4.2 vs.+1.9±5.2 mg/dL, P=0.01) and high-sensitivity C-reactive protein (hs-CRP) (−1.6±3.1 vs.+0.2±1.5 mg/L, P=0.004) compared to the placebo. We did not see any significant effect of flaxseed oil omega-3 supplementation on hormonal and other lipid profiles, and plasma nitric oxide levels.
Conclusions Overall, flaxseed oil omega-3 supplementation for 12 weeks in women with PCOS had beneficial effects on insulin metabolism, mF-G scores, serum triglycerides, VLDL-cholesterol and hs-CRP levels, but did not affect hormonal and other lipid profiles, and plasma nitric oxide levels.
Collapse
Affiliation(s)
- Golara Mirmasoumi
- Department of Biochemistry, Shahreza Branch, Islamic Azad University, Shahreza, Isfahan, I.R. Iran
| | - Mohammad Fazilati
- Department of Biochemistry, Shahreza Branch, Islamic Azad University, Shahreza, Isfahan, I.R. Iran
- Department of Biology, Pyamenoor University, Tehran, Iran
| | - Fatemeh Foroozanfard
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Vahedpoor
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Samaneh Mahmoodi
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Nazanin Esfeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Maryam Mohseni
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| |
Collapse
|
31
|
Increased expression of PGRN protein in follicular fluid and mRNA in granulosa cells in overweight patients with polycystic ovary syndrome. Eur J Obstet Gynecol Reprod Biol 2017; 218:106-112. [DOI: 10.1016/j.ejogrb.2017.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 09/07/2017] [Accepted: 09/16/2017] [Indexed: 12/22/2022]
|
32
|
Maktabi M, Chamani M, Asemi Z. The Effects of Vitamin D Supplementation on Metabolic Status of Patients with Polycystic Ovary Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Horm Metab Res 2017; 49:493-498. [PMID: 28679140 DOI: 10.1055/s-0043-107242] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Data on the effects of vitamin D supplementation on metabolic status of patients with polycystic ovary syndrome (PCOS) are scarce. The current study was conducted to evaluate the effects of vitamin D supplementation on metabolic status of patients with PCOS. This randomized double-blind, placebo-controlled trial was performed on 70 vitamin D-deficient (serum concentrations<20 ng/ml) women with phenotype B-PCOS according to the Rotterdam criteria aged 18-40 years old. Participants were randomly allocated into 2 groups to take either 50 000 IU vitamin D (n=35) or placebo (n=35) every 2 weeks for 12 weeks. Metabolic, endocrine, inflammation, and oxidative stress biomarkers were quantified at the beginning of the study and after 12-week intervention. After the 12-week intervention, compared to the placebo, vitamin D supplementation significantly decreased fasting plasma glucose (FPG) (-3.1±7.3 vs. +0.5±6.3 mg/dl, p=0.02), insulin (-1.4±3.6 vs. +2.6±7.0 μIU/ml, p=0.004), homeostasis model of assessment-estimated insulin resistance (-0.3±0.8 vs. +0.6±1.6, p=0.003), homeostasis model of assessment-estimated B cell function (-4.9±13.4 vs. +9.9±26.9, p=0.005), and increased quantitative insulin sensitivity check index (+0.01±0.01 vs. -0.02±0.05, p=0.007). Supplementation with vitamin D also led to significant reductions in serum high-sensitivity C-reactive protein (hs-CRP) (-0.7±1.4 vs. +0.5±2.1 μg/mL, p=0.009) and plasma malondialdehyde (MDA) levels (-0.1±0.5 vs. +0.9±2.1 μmol/l, p=0.01) compared to the placebo. Overall, vitamin D supplementation for 12 weeks in vitamin D-deficient women with phenotype B-PCOS had beneficial effects on glucose homeostasis parameters, hs-CRP, and MDA.
Collapse
Affiliation(s)
- Maryam Maktabi
- Department of Gynecology and Obstetrics, Endocrinology and Metabolism Research Center, School of Medicine, Arak University of Medical Sciences, Arak, I. R. Iran
| | - Maryam Chamani
- Department of Gynecology and Obstetrics, School of Medicine, Iran University of Medical Sciences, Tehran, I. R. Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I. R. Iran
| |
Collapse
|
33
|
Dambala K, Vavilis D, Bili E, Goulis DG, Tarlatzis BC. Serum visfatin, vascular endothelial growth factor and matrix metalloproteinase-9 in women with polycystic ovary syndrome. Gynecol Endocrinol 2017; 33:529-533. [PMID: 28300464 DOI: 10.1080/09513590.2017.1296425] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
AIM To evaluate serum concentrations of visfatin, vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP-9) in women with polycystic ovary syndrome (PCOS) and to investigate their possible role as early endothelial markers in PCOS. METHODS Forty-two women with PCOS and 42 controls, matched for age and weight, were included in the study. Serum concentrations of follicle-stimulating hormone (FSH), luteinizing hormone (LH), prolactin, total testosterone (tT), Δ4-androstenedione (Δ4A), dehydroepinadrosterone sulphate (DHEA-S), 17-OH-progesterone, sex hormone-binding protein (SHBG), thyroid-stimulating hormone (TSH), free thyroxine (fT4), visfatin, VEGF and MMP-9 were measured in all women; free androgen index (FAI) was calculated as well. Receiver-operating characteristic (ROC) analysis was performed to examine if visfatin, tT or FAI can predict the clinical status (PCOS or control). RESULTS LH, Δ4A, tT and FAI concentrations were higher in PCOS than in controls (p = 0.002, 0.029, 0.0005 and 0.014, respectively). Visfatin, VEGF and MMP-9 concentrations were higher in women with PCOS than controls (p = 0.019, 0.001 and 0.002, respectively). In ROC analysis, area under the curve (AUC) in the prediction of clinical status was 0.641 for visfatin (p = 0.026), 0.731 for tT (p = 0.001) and 0.666 for FAI (p = 0.010), with no difference among them (p = 0.117). CONCLUSIONS Visfatin may induce the expression of pro-angiogenic factors, such as VEGF and MMP-9, in women with PCOS, inplying gradually development of endothelial dysfunction. Further studies are required to clarify these findings.
Collapse
Affiliation(s)
- Kalliopi Dambala
- a First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Dimitrios Vavilis
- a First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Eleni Bili
- a First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Dimitrios G Goulis
- a First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Basil C Tarlatzis
- a First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki , Thessaloniki , Greece
| |
Collapse
|
34
|
VEJRAZKOVA D, LISCHKOVA O, VANKOVA M, STANICKA S, VRBIKOVA J, LUKASOVA P, VCELAK J, VACINOVA G, BENDLOVA B. Distinct Response of Fat and Gastrointestinal Tissue to Glucose in Gestational Diabetes Mellitus and Polycystic Ovary Syndrome. Physiol Res 2017; 66:283-292. [DOI: 10.33549/physiolres.933366] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gestational diabetes mellitus (GDM) and polycystic ovary syndrome (PCOS) are distinct pathologies with impaired insulin sensitivity as a common feature. The aim of this study was to evaluate the response of fat tissue adipokines and gastrointestinal incretins to glucose load in patients diagnosed with one of the two disorders and to compare it with healthy controls. Oral glucose tolerance test (oGTT) was performed in 77 lean young women: 22 had positive history of GDM, 19 were PCOS patients, and 36 were healthy controls. Hormones were evaluated in fasting and in 60 min intervals during the 3 h oGTT using Bio-Plex ProHuman Diabetes 10-Plex Assay for C-peptide, ghrelin, GIP, GLP1, glucagon, insulin, leptin, total PAI1, resistin, visfatin and Bio-Plex ProHuman Diabetes Adipsin and Adiponectin Assays (Bio-Rad). Despite lean body composition, both PCOS and GDM women were more insulin resistant than controls. Significant postchallenge differences between the GDM and PCOS groups were observed in secretion of adipsin, leptin, glucagon, visfatin, ghrelin, GIP, and also GLP1 with higher levels in GDM. Conversely, PCOS was associated with the highest resistin, C-peptide, and PAI1 levels. Our data suggest that decreased insulin sensitivity observed in lean women with GDM and PCOS is associated with distinct hormonal response of fat and gastrointestinal tissue to glucose load.
Collapse
Affiliation(s)
- D. VEJRAZKOVA
- Department of Molecular Endocrinology, Institute of Endocrinology, Prague, Czech Republic
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Karamali M, Ashrafi M, Razavi M, Jamilian M, Akbari M, Asemi Z. The Effects of Calcium, Vitamins D and K co-Supplementation on Markers of Insulin Metabolism and Lipid Profiles in Vitamin D-Deficient Women with Polycystic Ovary Syndrome. Exp Clin Endocrinol Diabetes 2017; 125:316-321. [PMID: 28407660 DOI: 10.1055/s-0043-104530] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Data on the effects of calcium, vitamins D and K co-supplementation on markers of insulin metabolism and lipid profiles among vitamin D-deficient women with polycystic ovary syndrome (PCOS) are scarce. This study was done to determine the effects of calcium, vitamins D and K co-supplementation on markers of insulin metabolism and lipid profiles in vitamin D-deficient women with PCOS. This randomized double-blind, placebo-controlled trial was conducted among 55 vitamin D-deficient women diagnosed with PCOS aged 18-40 years old. Subjects were randomly assigned into 2 groups to intake either 500 mg calcium, 200 IU vitamin D and 90 µg vitamin K supplements (n=28) or placebo (n=27) twice a day for 8 weeks. After the 8-week intervention, compared with the placebo, joint calcium, vitamins D and K supplementation resulted in significant decreases in serum insulin concentrations (-1.9±3.5 vs. +1.8±6.6 µIU/mL, P=0.01), homeostasis model of assessment-estimated insulin resistance (-0.4±0.7 vs. +0.4±1.4, P=0.01), homeostasis model of assessment-estimated b cell function (-7.9±14.7 vs. +7.0±30.3, P=0.02) and a significant increase in quantitative insulin sensitivity check index (+0.01±0.01 vs. -0.008±0.03, P=0.01). In addition, significant decreases in serum triglycerides (-23.4±71.3 vs. +9.9±39.5 mg/dL, P=0.03) and VLDL-cholesterol levels (-4.7±14.3 vs. +2.0±7.9 mg/dL, P=0.03) was observed following supplementation with combined calcium, vitamins D and K compared with the placebo. Overall, calcium, vitamins D and K co-supplementation for 8 weeks among vitamin D-deficient women with PCOS had beneficial effects on markers of insulin metabolism, serum triglycerides and VLDL-cholesterol levels.
Collapse
Affiliation(s)
- Maryam Karamali
- Department of Gynecology and Obstetrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahnaz Ashrafi
- Department of Gynecology and Obstetrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryamalsadat Razavi
- Department of Gynecology and Obstetrics, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mehri Jamilian
- Endocrinology and Metabolism Research Center, Department of Gynecology and Obstetrics, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Maryam Akbari
- Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| |
Collapse
|
36
|
Liu R, Zhang C, Shi Y, Zhang F, Li L, Wang X, Ling Y, Fu H, Dong W, Shen J, Reeves A, Greenberg AS, Zhao L, Peng Y, Ding X. Dysbiosis of Gut Microbiota Associated with Clinical Parameters in Polycystic Ovary Syndrome. Front Microbiol 2017; 8:324. [PMID: 28293234 PMCID: PMC5328957 DOI: 10.3389/fmicb.2017.00324] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/15/2017] [Indexed: 12/19/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder in women. Gut microbiota has been implicated to play a critical role in metabolic diseases and may modulate the secretion of mediators of the brain-gut axis. Interaction between gut microbiota and the endocrine and biochemical disturbances in PCOS still remains elusive. Here, we showed an altered gut microbiota significantly correlated with PCOS phenotype. There were 33 patients with PCOS (non-obese PCOS individuals, PN, n = 12; obese PCOS individuals, PO, n = 21) as well as 15 control subjects (non-obese control individuals, CN, n = 9; obese control individuals, CO, n = 6) enrolled in our study. The plasma levels of serotonin, ghrelin, and peptide YY (PYY) were significantly decreased in patients with PCOS compared with controls, and have a significantly negative correlation with waist circumference and testosterone. Sequencing of the V3-V4 region of the 16S rRNA gene in fecal samples revealed the substantial differences of gut microbial species between the PCOS and non-obese controls. Bacterial species were clustered into 23 co-abundance groups (CAGs) based on the SparCC correlation coefficients of their relative abundance. The CAGs increased in PCOS, including the bacteria belonging to Bacteroides, Escherichia/Shigella and Streptococcus, were negatively correlated with ghrelin, and positively correlated with testosterone and BMI. Furthermore, the CAGs that were decreased in PCOS, including the bacteria from Akkermansia and Ruminococcaceae, showed opposite relationship with body-weight, sex-hormone, and brain-gut peptides. In conclusion, gut microbial dysbiosis in women with PCOS is associated with the disease phenotypes.
Collapse
Affiliation(s)
- Rui Liu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong UniversityShanghai, China
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong UniversityShanghai, China
| | - Yu Shi
- Department of Endocrinology and Metabolism, Qidong People’s HospitalJiangsu, China
| | - Feng Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong UniversityShanghai, China
| | - Linxia Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Xuejiao Wang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Yunxia Ling
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Huaqing Fu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong UniversityShanghai, China
| | - Weiping Dong
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Jian Shen
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong UniversityShanghai, China
| | - Andrew Reeves
- Obesity and Metabolism Laboratory, Jean-Mayer USDA Human Nutrition Research Center on Aging at Tufts University, BostonMA, USA
| | - Andrew S. Greenberg
- Obesity and Metabolism Laboratory, Jean-Mayer USDA Human Nutrition Research Center on Aging at Tufts University, BostonMA, USA
| | - Liping Zhao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong UniversityShanghai, China
| | - Yongde Peng
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Xiaoying Ding
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| |
Collapse
|
37
|
Polak K, Czyzyk A, Simoncini T, Meczekalski B. New markers of insulin resistance in polycystic ovary syndrome. J Endocrinol Invest 2017; 40:1-8. [PMID: 27473078 PMCID: PMC5206255 DOI: 10.1007/s40618-016-0523-8] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 07/21/2016] [Indexed: 02/08/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine-metabolic disorder in women of reproductive age. The diagnostic criteria include two out of three features: hyperandrogenism, polycystic ovaries on ultrasound and menstrual irregularities (Rotterdam Criteria 2003). PCOS patients are more vulnerable to develop diabetes, cardiovascular diseases and metabolic syndrome. Insulin resistance (IR) is prevalent in women with PCOS independently of obesity and is critically involved in reproductive and metabolic complications of the syndrome. Several tests have been developed to measure IR, some very reliable but complex like the hyperinsulinemic euglycemic glucose clamp and others less precise but easier and less invasive like HOMA-IR. New markers are needed to reach a more reliable assessment of insulin resistance. To date, several surrogate markers have been proposed in the literature to facilitate and improve the determination of IR. Many new proteins are strongly involved with PCOS physiopathology and IR, such as some adipocytokines (adiponectin, visfatin, vaspin and apelin), copeptin, irisin, PAI-1 and zonulin. Many other proteins have been proposed as potential new markers of IR in PCOS, such as resistin, leptin, RBP4, kisspetin and ghrelin, but their role is still controversial. In this review, we provide a short characterization of these new markers, recently studied as indicators of metabolic state.
Collapse
Affiliation(s)
- K Polak
- Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology, University of Pisa, Pisa, Italy
| | - A Czyzyk
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, ul. Polna 33, Poznan, Poland
| | - T Simoncini
- Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology, University of Pisa, Pisa, Italy
| | - B Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, ul. Polna 33, Poznan, Poland.
| |
Collapse
|
38
|
Daan NMP, Koster MPH, de Wilde MA, Dalmeijer GW, Evelein AMV, Fauser BCJM, de Jager W. Biomarker Profiles in Women with PCOS and PCOS Offspring; A Pilot Study. PLoS One 2016; 11:e0165033. [PMID: 27806063 PMCID: PMC5091782 DOI: 10.1371/journal.pone.0165033] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 10/05/2016] [Indexed: 11/22/2022] Open
Abstract
Objective To study metabolic/inflammatory biomarker risk profiles in women with PCOS and PCOS offspring. Design Cross-sectional comparison of serum biomarkers. Setting University Medical Center Utrecht. Patients Hyperandrogenic PCOS women (HA-PCOS, n = 34), normoandrogenic PCOS women (NA-PCOS, n = 34), non-PCOS reference population (n = 32), PCOS offspring (n = 14, age 6–8 years), and a paedriatic reference population (n = 30). Main Outcome Measure(s) Clustering profile of adipocytokines (IL-1b, IL-6, IL-13, IL-17, IL-18, TNF-α, adiponectin, adipsin, leptin, chemerin, resistin, RBP4, DPP-IV/sCD26, CCL2/MCP-1), growth factors (PIGF, VEGF, sVEGF-R1), soluble cell adhesion molecules (sICAM-1/sCD54, sVCAM-1/sCD106), and other inflammatory related proteases (MMP-9, S100A8, Cathepsin S). Differences in median biomarker concentrations between groups, and associations with the free androgen index (FAI; Testosterone/SHBG x100). Results The cluster analysis identified leptin, RBP-4, DPP-IV and adiponectin as potential discriminative markers for HA-PCOS with a specifically strong correlation in cases with increased BMI. Leptin (R2 = 0.219) and adiponectin (R2 = 0.182) showed the strongest correlation with the FAI. When comparing median protein concentrations adult PCOS women with or without hyperandrogenemia, the most profound differences were observed for leptin (P < 0.001), DPP-IV (P = 0.005), and adiponectin (P < 0.001). Adjusting for age, BMI and multiple testing attenuated all differences. In PCOS offspring, MMP-9 (P = 0.001) and S100A8 (P < 0.001) concentrations were significantly higher compared to a healthy matched reference population, even after correcting for age and BMI and adjustment for multiple testing. Conclusion In this preliminary investigation we observed significant differences in adipocytokines between women with or without hyperandrogenic PCOS and non-PCOS controls, mostly influenced by BMI. Leptin and adiponectin showed the strongest correlation with the FAI in adult women with PCOS. In PCOS offspring other inflammatory biomarkers (MMP-9, S100A8) were increased, suggesting that these children may exhibit increased chronic low-grade inflammation. Additional research is required to confirm results of the current exploratory investigation.
Collapse
Affiliation(s)
- Nadine M. P. Daan
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
- * E-mail:
| | - Maria P. H. Koster
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marlieke A. de Wilde
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gerdien W. Dalmeijer
- Julius Centre for Health Sciences and Primary care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Annemieke M. V. Evelein
- Julius Centre for Health Sciences and Primary care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Bart C. J. M. Fauser
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wilco de Jager
- Laboratory of Translational Immunology, Division of Pediatrics, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| |
Collapse
|
39
|
Yildizhan B, Anik Ilhan G, Pekin T. The impact of insulin resistance on clinical, hormonal and metabolic parameters in lean women with polycystic ovary syndrome. J OBSTET GYNAECOL 2016; 36:893-896. [PMID: 27140180 DOI: 10.3109/01443615.2016.1168376] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This study was performed to assess insulin resistance (IR) in lean women with polycystic ovary syndrome (PCOS). Retrospective analysis of 100 consecutive lean (body mass index <25 kg/m2) PCOS subjects was performed. Subjects were divided into two groups according to homeostasis model assessment IR index (HOMA-IR), as IR + and IR-. A HOMA-IR value >2.5 was used to indicate IR. A total of 100 lean PCOS subjects were enrolled in the study, of which 47% were insulin resistant. Comparison of group means showed significantly higher values for waist-to-hip ratio (WHR), diastolic blood pressure and Ferriman-Gallwey score (FGS) in IR + group. HOMA-IR values were found to be positively correlated with WHR (r = 0.500, p < 0.01), systolic blood pressure (r = 0.265, p < 0.01), diastolic blood pressure (r = 0.273, p < 0.01), estradiol levels (r = 0.218, p < 0.05), FGS (r = 0.456, p < 0.01) and total testosterone levels (r = 0.291, p < 0.01). When evaluating PCOS subjects, the insulin resistant group should be separated as unique and IR should also be evaluated in lean women with PCOS.
Collapse
Affiliation(s)
- Begum Yildizhan
- a Department of Obstetrics and Gynecology , Marmara University , Istanbul , Turkey
| | - Gokce Anik Ilhan
- a Department of Obstetrics and Gynecology , Marmara University , Istanbul , Turkey
| | - Tanju Pekin
- a Department of Obstetrics and Gynecology , Marmara University , Istanbul , Turkey
| |
Collapse
|
40
|
Ding L, Gao F, Zhang M, Yan W, Tang R, Zhang C, Chen ZJ. Higher PDCD4 expression is associated with obesity, insulin resistance, lipid metabolism disorders, and granulosa cell apoptosis in polycystic ovary syndrome. Fertil Steril 2016; 105:1330-1337.e3. [DOI: 10.1016/j.fertnstert.2016.01.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/31/2015] [Accepted: 01/18/2016] [Indexed: 12/16/2022]
|
41
|
Gao T, Wu L, Chang F, Cao G. Low circulating ghrelin levels in women with polycystic ovary syndrome: a systematic review and meta-analysis. Endocr J 2016; 63:93-100. [PMID: 26607017 PMCID: PMC4975374 DOI: 10.1507/endocrj.ej15-0318] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although numerous, human subject studies evaluating the relationship between circulating ghrelin levels and polycystic ovary syndrome (PCOS) risk have yielded inconsistent findings. We aimed to quantitatively assess the association by summarizing all available evidence from human subject studies. The PubMed and Web of Science databases were searched up to February 2015 for eligible studies. Studies were eligible if they reported circulating ghrelin levels in women with PCOS and healthy women controls. A fixed or random-effects model was used to pool risk estimations. Twenty studies including 894 PCOS patients and 574 controls were included in the meta-analysis. The studies had fair methodological quality. The pooling analysis of all available studies revealed that ghrelin levels were significantly lower in PCOS patients than in controls, with standardized mean difference of -0.40 (95% CI: -0.73, -0.08). The significant association persisted in many subgroup strata. However, the heterogeneity across studies was considerable and not eliminated in subgroup analyses. Meta-regression analysis further suggested that the heterogeneity might be relevant to variability in study location, PCOS relevant factors like HOMA-IR ratio, as well as other factors not assessed. In conclusion, our meta-analysis suggested that ghrelin levels were significantly lower in PCOS patients than in controls. Further studies with large sample sizes are warranted to replicate our findings.
Collapse
Affiliation(s)
- Tian Gao
- Department of Veterinary, Inner Mongolia Agricultural University, Hohhot, MO, 010018, People’s Republic of China
- Center for safty evaluation of drugs, Inner Mongolia Medical University, Hohhot, MO, 010110, People’s Republic of China
| | - Lang Wu
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Fuhou Chang
- Center for safty evaluation of drugs, Inner Mongolia Medical University, Hohhot, MO, 010110, People’s Republic of China
| | - Guifang Cao
- Department of Veterinary, Inner Mongolia Agricultural University, Hohhot, MO, 010018, People’s Republic of China
| |
Collapse
|
42
|
Jamilian M, Razavi M, Fakhrie Kashan Z, Ghandi Y, Bagherian T, Asemi Z. Metabolic response to selenium supplementation in women with polycystic ovary syndrome: a randomized, double-blind, placebo-controlled trial. Clin Endocrinol (Oxf) 2015; 82:885-91. [PMID: 25510442 DOI: 10.1111/cen.12699] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 12/03/2014] [Accepted: 12/07/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE We are aware of no study examining the effects of selenium supplementation on metabolic profiles of patients with polycystic ovary syndrome (PCOS). This study was conducted to evaluate the effects of selenium supplementation on glucose homeostasis parameters and lipid concentrations in women with PCOS. DESIGN, PATIENTS AND MEASUREMENTS This randomized, double-blind, placebo-controlled trial was conducted among 70 women diagnosed with PCOS and aged 18-40 years old. Participants were randomly divided into two groups to receive 200 μg per day selenium supplements (N = 35) or placebo (N = 35) for 8 weeks. Fasting blood samples were taken at baseline and after 8 weeks intervention to quantify glucose, insulin and lipid concentrations. RESULTS After 8 weeks of intervention, subjects who received selenium supplements had significantly decreased serum insulin levels (-29·83 ± 47·29 vs +9·07 ± 77·12 pmol/l, P = 0·013), homeostasis model of assessment-insulin resistance (HOMA-IR) (-1·15 ± 1·81 vs +0·42 ± 3·09, P = 0·011), homeostatic model assessment-beta-cell function (HOMA-B) (-19·06 ± 30·95 vs +4·55 ± 47·99, P = 0·017) and increased quantitative insulin sensitivity check index (QUICKI) (+0·03 ± 0·04 vs +0·0009 ± 0·05, P = 0·032) compared with placebo. In addition, supplementation with selenium resulted in a significant reduction in serum triglycerides (-0·14 ± 0·55 vs +0·11 ± 0·30 mmol/l, P = 0·025) and VLDL-C concentrations (-0·03 ± 0·11 vs +0·02 ± 0·06 mmol/l, P = 0·025) compared with placebo. CONCLUSIONS In conclusion, 200 microgram per day selenium supplementation for 8 weeks among PCOS women had beneficial effects on insulin metabolism parameters, triglycerides and VLDL-C levels; however, it did not affect FPG and other lipid profiles.
Collapse
Affiliation(s)
- Mehri Jamilian
- Department of Gynecology and Obstetrics, School of Medicine, Arak University of Medical Sciences, Arak, I.R. Iran
| | - Maryamalsadat Razavi
- Department of Gynecology and Obstetrics, School of Medicine, Ardabil University of Medical Sciences, Ardabil, I.R. Iran
| | - Zohreh Fakhrie Kashan
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Yasser Ghandi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Tayebeh Bagherian
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| |
Collapse
|
43
|
Foroozanfard F, Jamilian M, Jafari Z, Khassaf A, Hosseini A, Khorammian H, Asemi Z. Effects of Zinc Supplementation on Markers of Insulin Resistance and Lipid Profiles in Women with Polycystic Ovary Syndrome: a Randomized, Double-blind, Placebo-controlled Trial. Exp Clin Endocrinol Diabetes 2015; 123:215-20. [DOI: 10.1055/s-0035-1548790] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- F. Foroozanfard
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - M. Jamilian
- Department of Gynecology and Obstetrics, School of Medicine, Arak University of Medical Sciences, Arak, I.R. Iran
| | - Z. Jafari
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - A. Khassaf
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - A. Hosseini
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - H. Khorammian
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Z. Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| |
Collapse
|