1
|
Luo L, Wang W, Xu Y, Yang Y, Zhang L, Gao J, Mai J, Wang Q, Gong F. Differences in preimplantation blastocyst chromosomal aberrations between polycystic ovary syndrome women and controls: a multi-center retrospective cohort study. J Assist Reprod Genet 2024; 41:3051-3059. [PMID: 39287709 PMCID: PMC11621251 DOI: 10.1007/s10815-024-03235-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/17/2024] [Indexed: 09/19/2024] Open
Abstract
PURPOSE Comprehensive chromosomal status of blastocyst from women with polycystic ovary syndrome (PCOS) was limited. This study aimed to identify possible differences in the preimplantation blastocyst chromosome aberrations between PCOS women and controls receiving preimplantation genetic testing (PGT). METHODS This was a multi-center retrospective cohort study including a total of 707 blastocysts from 147 PCOS women and 3006 blastocysts from 821 control women receiving PGT between 2015 and 2021. Embryonic chromosomal aberration spectrums were compared between PCOS and controls. Mixed effects generalized linear model was conducted to explore possible influence of PCOS-related endocrinological disorders on embryonic chromosomal abnormalities. RESULTS Blastocysts from PCOS demonstrated significantly lower aneuploidy rate (15.2% vs. 25.2% per women, P < 0.001; 14.7% vs. 25.4% per blastocyst, P < 0.001) but greater mosaicism rate (12.5% vs. 8.0% per women, P = 0.007; 16.5% vs. 8.7% per blastocyst, P < 0.001). Mixed effects generalized linear model identified PCOS as an independent protective factor against embryonic aneuploidy (adjusted odds ratio = 0.68, 95% confidence interval, 0.50-0.93, P = 0.014) but a risk factor for embryonic mosaicism (adjusted odds ratio = 1.52, 95% confidence interval 1.11-2.10, P = 0.009). Further model analysis suggested that insulin resistance could be responsible for the increased risk of embryonic mosaicism among PCOS women (adjusted odds ratio = 2.17, 95% confidence interval, 1.10-4.31, P = 0.026). CONCLUSION PCOS is associated with a lower aneuploidy risk but an increased mosaicism risk in preimplantation blastocysts, and insulin resistance should be investigated as a potential cause.
Collapse
Affiliation(s)
- Lu Luo
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, First Affiliated Hospital, Sun Yat-Sen University, Zhongshan Road 2, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Wenjun Wang
- Reproductive Medicine Center, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yan Xu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, First Affiliated Hospital, Sun Yat-Sen University, Zhongshan Road 2, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Yuanyuan Yang
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410083, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410083, China
| | - Limei Zhang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, First Affiliated Hospital, Sun Yat-Sen University, Zhongshan Road 2, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Jun Gao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, First Affiliated Hospital, Sun Yat-Sen University, Zhongshan Road 2, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Jiayi Mai
- Reproductive Medicine Center, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiong Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, First Affiliated Hospital, Sun Yat-Sen University, Zhongshan Road 2, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China.
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China.
| | - Fei Gong
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410083, China.
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410083, China.
| |
Collapse
|
2
|
Misra S, Gada J, Dhole C, Varthakavi P, Bhagwat N. Comparative Study of Insulin Sensitivity and Resistance and Their Correlation with Androgens in Lean and Obese Women with Polycystic Ovary Syndrome. Reprod Sci 2024; 31:754-763. [PMID: 37848646 DOI: 10.1007/s43032-023-01374-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/01/2023] [Indexed: 10/19/2023]
Abstract
There is a lack of consensus on the optimal screening strategy for insulin resistance (IR), particularly in lean women with polycystic ovary syndrome (PCOS). Therefore, we conducted a cross-sectional study in 80 women with PCOS (28 lean/52 obese) and 80 age- and body mass index (BMI)-matched controls. Using a 5-point 75-g oral glucose tolerance test (OGTT) (0, 30, 60, 90, 120 min), we examined glucose and insulin excursions, IR, insulin sensitivity, beta-cell function (ßF), and the effect of androgens on IR. Lean and obese women with PCOS had similar glucose but higher insulin (except fasting in lean women) and insulin AUC as compared to their respective controls (p < 0.05). Lean women with PCOS were equally insulin-resistant but more hyperinsulinemic than the obese controls (p < 0.05). Although ßF ([1st phase: 481.71 ± 263.53 vs. 430.56 ± 232.37], [2nd phase: 815.16 ± 447.12 vs. 752.66 ± 428.95]) was comparable in lean and obese women with PCOS, lean women had better insulin sensitivity (112.78 ± 66.26 vs. 75.49 ± 55.6) (p < 0.05). Dehydroepiandrosterone sulfate (DHEAS) and androstenedione decreased with increasing BMI in lean women, and this correlated with deteriorating insulin sensitivity and exaggerated hyperinsulinemia. In obese women with PCOS, sex hormone-binding globulin (SHBG) correlated negatively with BMI and hyperinsulinemia, and positively with insulin sensitivity. This data suggests that estimating only fasting insulin may miss IR in lean women with PCOS; hence, additional time points in OGTT will add value to screening for IR. DHEAS and androstenedione may have a beneficial effect on insulin sensitivity and may be used to screen IR in lean women, while SHBG can be used as a predictive marker for IR in obese women with PCOS.
Collapse
Affiliation(s)
- Sukirti Misra
- Department of Endocrinology, College Building, Topiwala National Medical College and Bai Yamunabai Laxman (BYL) Nair Charitable Hospital, A.L. Nair Road, Room No. 419, 4Th Floor, Mumbai Central, Mumbai, Maharashtra, 400008, India
| | - Jugal Gada
- Department of Endocrinology, College Building, Topiwala National Medical College and Bai Yamunabai Laxman (BYL) Nair Charitable Hospital, A.L. Nair Road, Room No. 419, 4Th Floor, Mumbai Central, Mumbai, Maharashtra, 400008, India.
| | - Charushila Dhole
- Department of Endocrinology, College Building, Topiwala National Medical College and Bai Yamunabai Laxman (BYL) Nair Charitable Hospital, A.L. Nair Road, Room No. 419, 4Th Floor, Mumbai Central, Mumbai, Maharashtra, 400008, India
| | - Premlata Varthakavi
- Department of Endocrinology, College Building, Topiwala National Medical College and Bai Yamunabai Laxman (BYL) Nair Charitable Hospital, A.L. Nair Road, Room No. 419, 4Th Floor, Mumbai Central, Mumbai, Maharashtra, 400008, India
| | - Nikhil Bhagwat
- Department of Endocrinology, College Building, Topiwala National Medical College and Bai Yamunabai Laxman (BYL) Nair Charitable Hospital, A.L. Nair Road, Room No. 419, 4Th Floor, Mumbai Central, Mumbai, Maharashtra, 400008, India
| |
Collapse
|
3
|
Burns K, Mullin BH, Moolhuijsen LME, Laisk T, Tyrmi JS, Cui J, Actkins KV, Louwers YV, Davis LK, Dudbridge F, Azziz R, Goodarzi MO, Laivuori H, Mägi R, Visser JA, Laven JSE, Wilson SG, Day FR, Stuckey BGA. Body mass index stratified meta-analysis of genome-wide association studies of polycystic ovary syndrome in women of European ancestry. BMC Genomics 2024; 25:208. [PMID: 38408933 PMCID: PMC10895801 DOI: 10.1186/s12864-024-09990-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 01/08/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a complex multifactorial disorder with a substantial genetic component. However, the clinical manifestations of PCOS are heterogeneous with notable differences between lean and obese women, implying a different pathophysiology manifesting in differential body mass index (BMI). We performed a meta-analysis of genome-wide association study (GWAS) data from six well-characterised cohorts, using a case-control study design stratified by BMI, aiming to identify genetic variants associated with lean and overweight/obese PCOS subtypes. RESULTS The study comprised 254,588 women (5,937 cases and 248,651 controls) from individual studies performed in Australia, Estonia, Finland, the Netherlands and United States of America, and separated according to three BMI stratifications (lean, overweight and obese). Genome-wide association analyses were performed for each stratification within each cohort, with the data for each BMI group meta-analysed using METAL software. Almost half of the total study population (47%, n = 119,584) were of lean BMI (≤ 25 kg/m2). Two genome-wide significant loci were identified for lean PCOS, led by rs12000707 within DENND1A (P = 1.55 × 10-12) and rs2228260 within XBP1 (P = 3.68 × 10-8). One additional locus, LINC02905, was highlighted as significantly associated with lean PCOS through gene-based analyses (P = 1.76 × 10-6). There were no significant loci observed for the overweight or obese sub-strata when analysed separately, however, when these strata were combined, an association signal led by rs569675099 within DENND1A reached genome-wide significance (P = 3.22 × 10-9) and a gene-based association was identified with ERBB4 (P = 1.59 × 10-6). Nineteen of 28 signals identified in previous GWAS, were replicated with consistent allelic effect in the lean stratum. There were less replicated signals in the overweight and obese groups, and only 4 SNPs were replicated in each of the three BMI strata. CONCLUSIONS Genetic variation at the XBP1, LINC02905 and ERBB4 loci were associated with PCOS within unique BMI strata, while DENND1A demonstrated associations across multiple strata, providing evidence of both distinct and shared genetic features between lean and overweight/obese PCOS-affected women. This study demonstrated that PCOS-affected women with contrasting body weight are not only phenotypically distinct but also show variation in genetic architecture; lean PCOS women typically display elevated gonadotrophin ratios, lower insulin resistance, higher androgen levels, including adrenal androgens, and more favourable lipid profiles. Overall, these findings add to the growing body of evidence supporting a genetic basis for PCOS as well as differences in genetic patterns relevant to PCOS BMI-subtype.
Collapse
Affiliation(s)
- Kharis Burns
- Department of Endocrinology and Diabetes, Royal Perth Hospital, Perth, WA, 6009, Australia.
- Medical School, University of Western Australia, Nedlands, WA, Australia.
| | - Benjamin H Mullin
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Loes M E Moolhuijsen
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Triin Laisk
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jaakko S Tyrmi
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Jinrui Cui
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ky'Era V Actkins
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yvonne V Louwers
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Lea K Davis
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Frank Dudbridge
- Population Health Sciences, University of Leicester, Leicester, UK
| | - Ricardo Azziz
- Obstetrics & Gynecology, Medicine, and Healthcare Organization & Policy, Schools of Medicine and Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hannele Laivuori
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland
- Institute for Molecular Medicine Finland, FIMM, hiLIFE, University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jenny A Visser
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joop S E Laven
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Scott G Wilson
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Felix R Day
- MRC Epidemiology Unit, Cambridge Biomedical Campus, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Bronwyn G A Stuckey
- Medical School, University of Western Australia, Nedlands, WA, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- Keogh Institute for Medical Research, Nedlands, WA, Australia
| |
Collapse
|
4
|
Bhattacharya K, Dey R, Sen D, Paul N, Basak AK, Purkait MP, Shukla N, Chaudhuri GR, Bhattacharya A, Maiti R, Adhikary K, Chatterjee P, Karak P, Syamal AK. Polycystic ovary syndrome and its management: In view of oxidative stress. Biomol Concepts 2024; 15:bmc-2022-0038. [PMID: 38242137 DOI: 10.1515/bmc-2022-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/11/2023] [Indexed: 01/21/2024] Open
Abstract
In the past two decades, oxidative stress (OS) has drawn a lot of interest due to the revelation that individuals with many persistent disorders including diabetes, polycystic ovarian syndrome (PCOS), cardiovascular, and other disorders often have aberrant oxidation statuses. OS has a close interplay with PCOS features such as insulin resistance, hyperandrogenism, and chronic inflammation; there is a belief that OS might contribute to the development of PCOS. PCOS is currently recognized as not only one of the most prevalent endocrine disorders but also a significant contributor to female infertility, affecting a considerable proportion of women globally. Therefore, the understanding of the relationship between OS and PCOS is crucial to the development of therapeutic and preventive strategies for PCOS. Moreover, the mechanistic study of intracellular reactive oxygen species/ reactive nitrogen species formation and its possible interaction with women's reproductive health is required, which includes complex enzymatic and non-enzymatic antioxidant systems. Apart from that, our current review includes possible regulation of the pathogenesis of OS. A change in lifestyle, including physical activity, various supplements that boost antioxidant levels, particularly vitamins, and the usage of medicinal herbs, is thought to be the best way to combat this occurrence of OS and improve the pathophysiologic conditions associated with PCOS.
Collapse
Affiliation(s)
- Koushik Bhattacharya
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Khurda Road, Bhubaneswar, Odisha, India
| | - Rajen Dey
- Department of Medical Laboratory Technology, Swami Vivekananda University, Barrackpore, West Bengal, India
| | - Debanjana Sen
- Post-Graduate Department of Physiology, Hooghly Mohsin College, Chinsurah, West-Bengal, India
| | - Nimisha Paul
- Department of General Human Physiology and Biochemistry, Hitkarini Dental College and Hospital, Jabalpur, Madhya Pradesh, India
| | - Asim Kumar Basak
- School of Allied Health Sciences, Brainware University, Barasat, West-Bengal, India
| | | | - Nandini Shukla
- Department of Anatomy, Pt. J.N.M. Medical College, Raipur, Chhattisgarh, India
| | - Gargi Ray Chaudhuri
- Department of Physiotherapy, Nopany Institute of Health Care Studies, Kolkata, West-Bengal, India
| | - Aniruddha Bhattacharya
- Department of Physiology, International Medical School, Management and Science University, Selangor, Malaysia
| | - Rajkumar Maiti
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, India
| | - Krishnendu Adhikary
- Department of Interdisciplinary Science, Centurion University of Technology and Management, Khurda Road, Bhubaneswar, Odisha, India
| | - Prity Chatterjee
- Department of Biotechnology, Paramedical College, Durgapur, West Bengal, India
| | - Prithviraj Karak
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, India
| | - Alak Kumar Syamal
- Post-Graduate Department of Physiology, Hooghly Mohsin College, Chinsurah, West-Bengal, India
| |
Collapse
|
5
|
Zhu X, Liu L, Feng Z, Zhang Y. Correlation of plasma adipokines with endometrial atypical hyperplasia and type I/II endometrial cancer. J OBSTET GYNAECOL 2023; 43:2179914. [PMID: 36815556 DOI: 10.1080/01443615.2023.2179914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
The aim of the study was to systematically explore the relationships between various adipokines and risks of endometrial atypical hyperplasia (EAH), type I endometrial cancer (EC), and type II EC. We enrolled 219 patients in this study, including 39 EAH, 87 type I EC, 38 type II EC and 55 control individuals. We subsequently explored the association of adipokine levels and the leptin-to-adiponectin (L/A) ratio with EAH, type I EC, and type II EC. The plasma leptin level and L/A ratio were significantly higher in the EAH group than in the control group (p = 0.012). Leptin, resistin, vaspin, and visfatin levels were significantly higher in the type I EC group; however, the adiponectin level was lower in the type I EC, which resulted in a higher L/A ratio. Notably, the L/A ratio and visfatin level in the type II EC group were significantly higher. Multiple logistic regression analysis revealed that a higher leptin level was significantly associated with a higher EAH risk (p = 0.012). Higher leptin level (p = 0.042) and L/A ratio (p = 0.027) were significantly associated with an increased type I EC risk. By contrast, higher leptin (p = 0.059) and visfatin (p = 0.003) levels, higher L/A ratio (p = 0.033), and lower adiponectin level (p = 0.042) were associated with an increased type II EC risk. We suggested that adipokines are potentially correlated with EAH and EC risks.
Collapse
Affiliation(s)
- Xinxin Zhu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Linzhi Liu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Zonghao Feng
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| |
Collapse
|
6
|
Parker J. Pathophysiological Effects of Contemporary Lifestyle on Evolutionary-Conserved Survival Mechanisms in Polycystic Ovary Syndrome. Life (Basel) 2023; 13:life13041056. [PMID: 37109585 PMCID: PMC10145572 DOI: 10.3390/life13041056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is increasingly being characterized as an evolutionary mismatch disorder that presents with a complex mixture of metabolic and endocrine symptoms. The Evolutionary Model proposes that PCOS arises from a collection of inherited polymorphisms that have been consistently demonstrated in a variety of ethnic groups and races. In utero developmental programming of susceptible genomic variants are thought to predispose the offspring to develop PCOS. Postnatal exposure to lifestyle and environmental risk factors results in epigenetic activation of developmentally programmed genes and disturbance of the hallmarks of health. The resulting pathophysiological changes represent the consequences of poor-quality diet, sedentary behaviour, endocrine disrupting chemicals, stress, circadian disruption, and other lifestyle factors. Emerging evidence suggests that lifestyle-induced gastrointestinal dysbiosis plays a central role in the pathogenesis of PCOS. Lifestyle and environmental exposures initiate changes that result in disturbance of the gastrointestinal microbiome (dysbiosis), immune dysregulation (chronic inflammation), altered metabolism (insulin resistance), endocrine and reproductive imbalance (hyperandrogenism), and central nervous system dysfunction (neuroendocrine and autonomic nervous system). PCOS can be a progressive metabolic condition that leads to obesity, gestational diabetes, type two diabetes, metabolic-associated fatty liver disease, metabolic syndrome, cardiovascular disease, and cancer. This review explores the mechanisms that underpin the evolutionary mismatch between ancient survival pathways and contemporary lifestyle factors involved in the pathogenesis and pathophysiology of PCOS.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
7
|
Berbrier DE, Leone CA, Adler TE, Bender JR, Taylor HS, Stachenfeld NS, Usselman CW. Effects of androgen excess and body mass index on endothelial function in women with polycystic ovary syndrome. J Appl Physiol (1985) 2023; 134:868-878. [PMID: 36861670 DOI: 10.1152/japplphysiol.00583.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is associated with endothelial dysfunction; whether this is attributable to comorbid hyperandrogenism and/or obesity remains to be established. Therefore, we 1) compared endothelial function between lean and overweight/obese (OW/OB) women with and without androgen excess (AE)-PCOS and 2) examined androgens as potential modulators of endothelial function in these women. The flow-mediated dilation (FMD) test was applied in 14 women with AE-PCOS (lean: n = 7; OW/OB: n = 7) and 14 controls (CTRL; lean: n = 7, OW/OB: n = 7) at baseline (BSL) and following 7 days of ethinyl estradiol supplementation (EE; 30 µg/day) to assess the effect of a vasodilatory therapeutic on endothelial function; at each time point we assessed peak increases in diameter during reactive hyperemia (%FMD), shear rate, and low flow-mediated constriction (%LFMC). BSL %FMD was attenuated in lean AE-PCOS versus both lean CTRL (5.2 ± 1.5 vs. 10.3 ± 2.6%, P < 0.01) and OW/OB AE-PCOS (5.2 ± 1.5 vs. 6.6 ± 0.9%, P = 0.048). A negative correlation between BSL %FMD and free testosterone was observed in lean AE-PCOS only (R2 = 0.68, P = 0.02). EE increased %FMD in both OW/OB groups (CTRL: 7.6 ± 0.6 vs. 10.4 ± 2.5%, AE-PCOS: 6.6 ± 0.9 vs. 9.6 ± 1.7%, P < 0.01), had no impact on %FMD in lean AE-PCOS (5.17 ± 1.5 vs. 5.17 ± 1.1%, P = 0.99), and reduced %FMD in lean CTRL (10.3 ± 2.6 vs. 7.6 ± 1.2%, P = 0.03). Collectively, these data indicate that lean women with AE-PCOS exhibit more severe endothelial dysfunction than their OW/OB counterparts. Furthermore, endothelial dysfunction appears to be mediated by circulating androgens in lean but not in OW/OB AE-PCOS, suggesting a difference in the endothelial pathophysiology of AE-PCOS between these phenotypes.NEW & NOTEWORTHY We present evidence for marked endothelial dysfunction in lean women with androgen excess polycystic ovary syndrome (AE-PCOS) that is 1) associated with free testosterone levels, 2) impaired relative to overweight/obese women with AE-PCOS, and 3) unchanged following short-term ethinyl estradiol supplementation. These data indicate an important direct effect of androgens on the vascular system in women with AE-PCOS. Our data also suggest that the relationship between androgens and vascular health differs between phenotypes of AE-PCOS.
Collapse
Affiliation(s)
- Danielle E Berbrier
- Cardiovascular Health and Autonomic Regulation Laboratory, Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
| | - Cheryl A Leone
- The John B. Pierce Laboratory, Yale School of Medicine, New Haven, Connecticut, United States
| | - Tessa E Adler
- Cardiovascular Health and Autonomic Regulation Laboratory, Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
- The John B. Pierce Laboratory, Yale School of Medicine, New Haven, Connecticut, United States
| | - Jeffrey R Bender
- Departments of Internal Medicine (Cardiovascular Medicine) and Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, Connecticut, United States
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States
| | - Nina S Stachenfeld
- The John B. Pierce Laboratory, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States
| | - Charlotte W Usselman
- Cardiovascular Health and Autonomic Regulation Laboratory, Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
- The John B. Pierce Laboratory, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States
- McGill Research Centre for Physical Activity and Health, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
8
|
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: 35] [Impact Index Per Article: 11.7] [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
|
9
|
Parker J, O’Brien C, Hawrelak J, Gersh FL. Polycystic Ovary Syndrome: An Evolutionary Adaptation to Lifestyle and the Environment. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19031336. [PMID: 35162359 PMCID: PMC8835454 DOI: 10.3390/ijerph19031336] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023]
Abstract
Polycystic ovary syndrome (PCOS) is increasingly recognized as a complex metabolic disorder that manifests in genetically susceptible women following a range of negative exposures to nutritional and environmental factors related to contemporary lifestyle. The hypothesis that PCOS phenotypes are derived from a mismatch between ancient genetic survival mechanisms and modern lifestyle practices is supported by a diversity of research findings. The proposed evolutionary model of the pathogenesis of PCOS incorporates evidence related to evolutionary theory, genetic studies, in utero developmental epigenetic programming, transgenerational inheritance, metabolic features including insulin resistance, obesity and the apparent paradox of lean phenotypes, reproductive effects and subfertility, the impact of the microbiome and dysbiosis, endocrine-disrupting chemical exposure, and the influence of lifestyle factors such as poor-quality diet and physical inactivity. Based on these premises, the diverse lines of research are synthesized into a composite evolutionary model of the pathogenesis of PCOS. It is hoped that this model will assist clinicians and patients to understand the importance of lifestyle interventions in the prevention and management of PCOS and provide a conceptual framework for future research. It is appreciated that this theory represents a synthesis of the current evidence and that it is expected to evolve and change over time.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong 2500, Australia
- Correspondence:
| | - Claire O’Brien
- Faculty of Science and Technology, University of Canberra, Bruce 2617, Australia;
| | - Jason Hawrelak
- College of Health and Medicine, University of Tasmania, Hobart 7005, Australia;
| | - Felice L. Gersh
- College of Medicine, University of Arizona, Tucson, AZ 85004, USA;
| |
Collapse
|
10
|
Livadas S, Anagnostis P, Bosdou JK, Bantouna D, Paparodis R. Polycystic ovary syndrome and type 2 diabetes mellitus: A state-of-the-art review. World J Diabetes 2022; 13:5-26. [PMID: 35070056 PMCID: PMC8771268 DOI: 10.4239/wjd.v13.i1.5] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/30/2021] [Accepted: 12/25/2021] [Indexed: 02/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) often coexists with a wide spectrum of dysglycemic conditions, ranging from impaired glucose tolerance to type 2 diabetes mellitus (T2D), which occur to a greater extent compared to healthy body mass index-matched women. This concurrence of disorders is mainly attributed to common pathogenetic pathways linking the two entities, such as insulin resistance. However, due to methodological flaws in the available studies and the multifaceted nature of the syndrome, there has been substantial controversy as to the exact association between T2D and PCOS which has not yet been elucidated. The aim of this review is to present the best available evidence regarding the epidemiology of dysglycemia in PCOS, the unique pathophysiological mechanisms underlying the progression of dysglycemia, the most appropriate methods for assessing glycemic status and the risk factors for T2D development in this population, as well as T2D risk after transition to menopause. Proposals for application of a holistic approach to enable optimal management of T2D risk in PCOS are also provided. Specifically, adoption of a healthy lifestyle with adherence to improved dietary patterns, such the Mediterranean diet, avoidance of consumption of endocrine-disrupting foods and beverages, regular exercise, and the effect of certain medications, such as metformin and glucagon-like peptide 1 receptor agonists, are discussed. Furthermore, the maintenance of a healthy weight is highlighted as a key factor in achievement of a significant reduction of T2D risk in women with PCOS.
Collapse
Affiliation(s)
| | - Panagiotis Anagnostis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki 54636, Greece
| | - Julia K Bosdou
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynaecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki 54636, Greece
| | - Dimitra Bantouna
- Department of Pathology and Cytology, University of Patras School of Medicine, Patras 10563, Greece
| | - Rodis Paparodis
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 23456, United States
| |
Collapse
|
11
|
Nizigiyimana P, Xu B, Liu L, Luo L, Liu T, Jiang M, Liu Z, Li C, Luo X, Lei M. Gut microbiota is associated with differential metabolic characteristics: A study on a defined cohort of Africans and Chinese. Front Endocrinol (Lausanne) 2022; 13:942383. [PMID: 36246928 PMCID: PMC9554505 DOI: 10.3389/fendo.2022.942383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE This study intended to determine the associations between gut microbiota and glucose response in healthy individuals and analyze the connection between the gut microbiome and glucose-metabolism-related parameters. METHODS Fecal bacterial composition and anthropometric, body composition, body fat distribution, and biochemical measures were analyzed. A 75-g oral glucose tolerance test (OGTT) was given to each participant to investigate changes in glucagon-like peptide 1 (GLP-1), insulin, and glucose. The whole body fat and the regions of interest of local body composition were analyzed using dual-energy X-ray absorptiometry (DEXA), and gut microbiota composition was assessed through variable regions (V3-V4) of the bacterial 16s ribosomal RNA gene using high-throughput sequencing techniques. Spearman correlation analysis was used to evaluate the association between gut microbiota and clinical and metabolic changes. RESULTS The number of operational taxonomic units (OTUs) demonstrated a reduction in the diversity and composition of gut microbiota associated with enhanced adiposity, dyslipidemia, insulin resistance, and hyperglycemia. The alpha diversity revealed that microbiota diversity, richness, and composition were higher in the African group and lower in the Chinese group. Principal coordinates analysis (PCoA) plots of beta diversity showed significant variability in gut microbial community structure between the two groups (p = 0.0009). LEfSe analysis showed that phylum Bacteroidetes was significantly more abundant in the Chinese group, and this group also harbored members of the order Bacteroidales, family Bacteroidaceae, and genus Bacteroides. In contrast, the phylum Verrucomicrobia was significantly more prevalent in the African group (all p < 0.05). Concerning species, metastats analysis revealed 8 species in the Chinese group and 18 species in the African group that were significantly abundant. Spearman's correlation analysis demonstrated that gut microbiota correlated with the factors that related to glucose metabolism. CONCLUSION Our data suggest that there is an interaction between gut microbiota, host physiology, and glucometabolic pathways, and this could contribute to adiposity and pathophysiology of hyperlipidemia, insulin resistance, and hyperglycemia. These findings provide an important basis for determining the relation between the gut microbiota and the pathogenesis of various metabolic disorders.
Collapse
Affiliation(s)
- Paul Nizigiyimana
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Boya Xu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Lerong Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Liping Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Tingting Liu
- Department of Endocrinology, Haikou Hospital Affiliated to Xiangya School of Medicine, Central South University, Haikou, China
| | - Meng Jiang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Zehao Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Minxiang Lei
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Minxiang Lei,
| |
Collapse
|
12
|
Kim N, Chun S. Association between the serum estrone-to-estradiol ratio and parameters related to glucose metabolism and insulin resistance in women with polycystic ovary syndrome. Clin Exp Reprod Med 2021; 48:374-379. [PMID: 34875745 PMCID: PMC8651759 DOI: 10.5653/cerm.2021.04553] [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: 04/28/2021] [Accepted: 08/27/2021] [Indexed: 11/10/2022] Open
Abstract
Objective We aimed to evaluate associations between the ratio of serum estrone (E1) to estradiol (E2) and parameters related to serum glucose metabolism and insulin resistance in women with polycystic ovary syndrome (PCOS). Methods In total, 133 women between the ages of 18 and 35 diagnosed with PCOS were enrolled in this study. All participants with PCOS underwent blood tests to determine hormonal and biochemical metabolic parameters and a standard 2-hour 75-g oral glucose tolerance test. They were divided into two groups according to the serum E1-to-E2 ratio: group 1 (E1/E2 ratio <2.0) and group 2 (E1/E2 ratio ≥2.0). Results In the comparative analysis, the waist-to-hip ratio (WHR) was the only clinical variable that was significantly different between the two groups. Patients with a higher E1/E2 ratio showed higher fasting insulin levels, homeostasis model for insulin resistance, and postprandial glucose level at 2 hours (PPG2). In a correlation analysis, only PPG2 was significantly related to the serum E1/E2 ratio. However, after controlling for the confounding effects of body mass index (BMI) and WHR, fasting glucose was also significantly correlated with the serum E1/E2 ratio. Conclusion Women with PCOS with a higher serum E1/E2 ratio were found to be more likely to show higher fasting insulin and postprandial glucose levels. Significant correlations were found between the serum E1/E2 ratio and both fasting and postprandial serum glucose levels after adjusting for BMI and WHR in women with PCOS.
Collapse
Affiliation(s)
- Nayoung Kim
- Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Sungwook Chun
- Department of Obstetrics and Gynecology, Inje University Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| |
Collapse
|
13
|
Resveratrol and Markers of Polycystic Ovary Syndrome: a Systematic Review of Animal and Clinical Studies. Reprod Sci 2021; 29:2477-2487. [PMID: 34312768 DOI: 10.1007/s43032-021-00653-9] [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: 03/11/2021] [Accepted: 06/03/2021] [Indexed: 10/20/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common disorder affecting childbearing-age women, and is associated with reproductive and metabolic disturbances. The present study aimed to systematically review current animal studies and randomized placebo-controlled clinical trials (RCT) regarding the effects of resveratrol, a natural polyphenolic compound, on PCOS features. PubMed, Scopus, Web of Knowledge, and Google Scholar were comprehensively searched until December 2020. All original animal articles and RCTs evaluating the effects of resveratrol on PCOS were eligible for the review. Out of 289 initial records, eight animal studies and three RCTs met our inclusion criteria. Most of the included animal studies reported beneficial effects of resveratrol on the histomorphological features, sex hormones and gonadotropins, glycemic control, inflammation, and oxidative stress. Resveratrol also ameliorated ovarian volume, high-quality oocyte rate, high-quality embryo rate, androgens and gonadotropins concentrations, angiogenic factors levels, and endoplasmic reticulum stress in PCOS patients. Upregulation of sirtuin-1 was an examined mechanism proposed for some observed effects of resveratrol. The current literature is limited to conclude the beneficial effects of resveratrol on the management of PCOS. Although, according to the promising results of the animal studies and limited RCTs, resveratrol might be an effective phytochemical in PCOS control, especially regarding hormonal and reproductive abnormalities. More mechanistic studies and RCTs are warranted to obvious whether resveratrol can be prescribed in the clinical situation.
Collapse
|
14
|
Wang H, Zhang Y, Fang X, Kwak-Kim J, Wu L. Insulin Resistance Adversely Affect IVF Outcomes in Lean Women Without PCOS. Front Endocrinol (Lausanne) 2021; 12:734638. [PMID: 34552564 PMCID: PMC8450607 DOI: 10.3389/fendo.2021.734638] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/18/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To investigate the effects of insulin resistance (IR) on IVF outcomes and a potential underlying mechanism in lean women without PCOS. DESIGN A prospective cohort study at the University Clinic. SETTING IVF center at the University setting. PATIENTS A total of 155 lean women (body mass index <25) without PCOS undergoing IVF cycle. INTERVENTION Patients were allocated to IR and non-IR groups based on HOMA-M120. MAIN OUTCOME MEASURES IVF outcomes, including egg quality, the percentage of mature oocytes, fertilization rate, blastocyst formation rate, advanced embryo rate, and cumulative live birth rate were investigated. Auto-immune parameters, peripheral blood immunophenotypes, thyroid hormone, homocysteine, and 25-OH-vitamin D3 (25-OH-VD3) levels were analyzed. RESULTS The percentage of mature oocytes and blastocyst formation rate were significantly lower in the IR group as compared with those of the non-IR group (p<0.05, respectively). The proportion of peripheral blood CD19+ B cells was significantly higher in the IR group than those of the non-IR group (p<0.05). Homocysteine, 25-OH-VD3, and auto-immune parameters were the same between the two groups. CONCLUSION In lean infertile women without PCOS, IR is associated with the decreased percentage of mature eggs and poor embryo quality in which B cell immunity may play a role.
Collapse
Affiliation(s)
- Haoyu Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xuhui Fang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL, United States
- Center for Cancer Cell Biology, Immunology and Infection Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- *Correspondence: Joanne Kwak-Kim, ; Li Wu,
| | - Li Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Joanne Kwak-Kim, ; Li Wu,
| |
Collapse
|
15
|
Dapas M, Lin FTJ, Nadkarni GN, Sisk R, Legro RS, Urbanek M, Hayes MG, Dunaif A. Distinct subtypes of polycystic ovary syndrome with novel genetic associations: An unsupervised, phenotypic clustering analysis. PLoS Med 2020; 17:e1003132. [PMID: 32574161 PMCID: PMC7310679 DOI: 10.1371/journal.pmed.1003132] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common, complex genetic disorder affecting up to 15% of reproductive-age women worldwide, depending on the diagnostic criteria applied. These diagnostic criteria are based on expert opinion and have been the subject of considerable controversy. The phenotypic variation observed in PCOS is suggestive of an underlying genetic heterogeneity, but a recent meta-analysis of European ancestry PCOS cases found that the genetic architecture of PCOS defined by different diagnostic criteria was generally similar, suggesting that the criteria do not identify biologically distinct disease subtypes. We performed this study to test the hypothesis that there are biologically relevant subtypes of PCOS. METHODS AND FINDINGS Using biochemical and genotype data from a previously published PCOS genome-wide association study (GWAS), we investigated whether there were reproducible phenotypic subtypes of PCOS with subtype-specific genetic associations. Unsupervised hierarchical cluster analysis was performed on quantitative anthropometric, reproductive, and metabolic traits in a genotyped cohort of 893 PCOS cases (median and interquartile range [IQR]: age = 28 [25-32], body mass index [BMI] = 35.4 [28.2-41.5]). The clusters were replicated in an independent, ungenotyped cohort of 263 PCOS cases (median and IQR: age = 28 [24-33], BMI = 35.7 [28.4-42.3]). The clustering revealed 2 distinct PCOS subtypes: a "reproductive" group (21%-23%), characterized by higher luteinizing hormone (LH) and sex hormone binding globulin (SHBG) levels with relatively low BMI and insulin levels, and a "metabolic" group (37%-39%), characterized by higher BMI, glucose, and insulin levels with lower SHBG and LH levels. We performed a GWAS on the genotyped cohort, limiting the cases to either the reproductive or metabolic subtypes. We identified alleles in 4 loci that were associated with the reproductive subtype at genome-wide significance (PRDM2/KAZN, P = 2.2 × 10-10; IQCA1, P = 2.8 × 10-9; BMPR1B/UNC5C, P = 9.7 × 10-9; CDH10, P = 1.2 × 10-8) and one locus that was significantly associated with the metabolic subtype (KCNH7/FIGN, P = 1.0 × 10-8). We developed a predictive model to classify a separate, family-based cohort of 73 women with PCOS (median and IQR: age = 28 [25-33], BMI = 34.3 [27.8-42.3]) and found that the subtypes tended to cluster in families and that carriers of previously reported rare variants in DENND1A, a gene that regulates androgen biosynthesis, were significantly more likely to have the reproductive subtype of PCOS. Limitations of our study were that only PCOS cases of European ancestry diagnosed by National Institutes of Health (NIH) criteria were included, the sample sizes for the subtype GWAS were small, and the GWAS findings were not replicated. CONCLUSIONS In conclusion, we have found reproducible reproductive and metabolic subtypes of PCOS. Furthermore, these subtypes were associated with novel, to our knowledge, susceptibility loci. Our results suggest that these subtypes are biologically relevant because they appear to have distinct genetic architecture. This study demonstrates how phenotypic subtyping can be used to gain additional insights from GWAS data.
Collapse
Affiliation(s)
- Matthew Dapas
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Frederick T. J. Lin
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Girish N. Nadkarni
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ryan Sisk
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Richard S. Legro
- Department of Obstetrics and Gynecology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Margrit Urbanek
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Center for Reproductive Science, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - M. Geoffrey Hayes
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Department of Anthropology, Northwestern University, Evanston, Illinois, United States of America
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
16
|
Patel V, Menezes H, Menezes C, Bouwer S, Bostick-Smith CA, Speelman DL. Regular Mindful Yoga Practice as a Method to Improve Androgen Levels in Women With Polycystic Ovary Syndrome: A Randomized, Controlled Trial. J Osteopath Med 2020; 120:2764759. [PMID: 32285088 DOI: 10.7556/jaoa.2020.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Holistic approaches are needed to complement existing therapies for polycystic ovary syndrome (PCOS), a common disorder affecting the health of reproductive-aged females. OBJECTIVE To determine whether thrice-weekly mindful yoga practice improves endocrine, cardiometabolic, or psychological parameters in women with PCOS. METHODS Thirty-one women with PCOS between the ages of 23 and 42 years and living in Erie County, Pennsylvania, were recruited for this randomized, controlled study arm, which was part of a larger 3-part investigation. Women were randomly assigned to either a mindful yoga intervention group or no intervention (control) group. Group classes were 1 hour, thrice weekly. Initial endocrine, cardiometabolic, and psychological measurements were compared with measurements taken after the 3-month intervention period. Measurements included free testosterone, dehydroepiandrosterone, androstenedione, body mass index, waist-to-hip ratio, fasting blood glucose and insulin levels, and anxiety and depression scores. RESULTS Twenty-two women completed the 3-month intervention period, 13 in the mindful yoga group and 9 in the control group. Paired comparisons of pre- and postintervention parameters indicated that women who completed the mindful yoga intervention had significantly lower free testosterone levels (5.96 vs 4.24 pg/mL; P<.05) and dehydroepiandrosterone levels that trended lower. Improved testosterone may persist for several months after completion of a 3-month, thrice-weekly mindful yoga intervention. Additionally, improvements were seen in measures of anxiety and depression. CONCLUSION The improvements observed suggest that regular mindful yoga practice can be a useful complementary therapeutic option for women with PCOS, particularly for improving serum androgen levels, a hallmark feature of PCOS. This improvement occurred in the absence of weight loss and may persist even if there is a lapse in practice. (ClinicalTrials.gov No. NCT03383484).
Collapse
|
17
|
Chappell NR, Zhou B, Schutt AK, Gibbons WE, Blesson CS. Prenatal androgen induced lean PCOS impairs mitochondria and mRNA profiles in oocytes. Endocr Connect 2020; 9:EC-19-0553.R1. [PMID: 32101528 PMCID: PMC7159265 DOI: 10.1530/ec-19-0553] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/25/2020] [Indexed: 01/13/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the most common ovulatory defect in women. Although most PCOS patients are obese, a subset of PCOS women are lean but show similar risks for adverse fertility outcomes. A lean PCOS mouse model was created using prenatal androgen administration. This developmentally programmed mouse model was used for this study. Our objective was to investigate if mitochondrial structure and functions were compromised in oocytes obtained from lean PCOS mouse. The lean PCOS mouse model was validated by performing glucose tolerance test, HbA1c levels, body weight and estrous cycle analyses. Oocytes were isolated and were used to investigate inner mitochondrial membrane potential, oxidative stress, lipid peroxidation, ATP production, mtDNA copy number, transcript abundance, histology and electron microscopy. Our results demonstrate that lean PCOS mice has similar weight to that of the controls but exhibited glucose intolerance and hyperinsulinemia along with dysregulated estrus cycle. Analysis of their oocytes show impaired inner mitochondrial membrane function, elevated reactive oxygen species (ROS), increased RNA transcript abundance and aberrant ovarian histology. Electron microscopy of the oocytes showed impaired mitochondrial ultrastructure. In conclusion, the lean PCOS mouse model shows a decreased oocyte quality related to impaired mitochondrial ultrastructure and function.
Collapse
Affiliation(s)
- Neil R Chappell
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children’s Hospital, Houston, Texas, USA
| | - Beth Zhou
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children’s Hospital, Houston, Texas, USA
| | - Amy K Schutt
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children’s Hospital, Houston, Texas, USA
| | - William E Gibbons
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children’s Hospital, Houston, Texas, USA
| | - Chellakkan S Blesson
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
18
|
Ortiz-Flores AE, Luque-Ramírez M, Fernández-Durán E, Alvarez-Blasco F, Escobar-Morreale HF. Diagnosis of disorders of glucose tolerance in women with polycystic ovary syndrome (PCOS) at a tertiary care center: fasting plasma glucose or oral glucose tolerance test? Metabolism 2019; 93:86-92. [PMID: 30710572 DOI: 10.1016/j.metabol.2019.01.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/22/2019] [Accepted: 01/26/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The risk of developing prediabetes and type 2 diabetes (dysglycemia) may be increased in women with PCOS. Whether an oral glucose tolerance test (OGTT) should be performed routinely in all PCOS women at presentation or should be recommended only to a selected subset of patients is still controversial. BASIC PROCEDURES At a tertiary care center, we conducted a retrospective, observational study including 400 women with PCOS submitted to an OGTT. Our primary objective was to assess the diagnostic agreement between two algorithms commonly used for the screening of dysglycemia in these women: i) relying only on fasting plasma glucose (FPG) or ii) considering both fasting and/or 120-min plasma glucose concentrations during an OGTT. We conducted the analysis considering all patients as a whole, and also after stratifying them by body weight, androgen concentrations and age. MAIN FINDINGS The OGTT detected dysglycemia in 24.5% of patients, whereas only 14.3% women would have been diagnosed using FPG levels alone. The latter missed as many as 40% of women with dysglycemia in our series, including all cases of diabetes. Diagnostic agreement between both algorithms was only 0.55 (κ = 0.103; 95% CI: 0.05-0.16). Areas under the receiver operating characteristic curve for dysglycemia were 0.86 (95%CI: 0.81-0.91) for FPG and 0.91 (95%CI = 0.87-0.95) for 120-min plasma glucose during the OGTT. FPG was not accurate in predicting dysglycemia in women with PCOS regardless of the presence of insulin resistance, weight excess, hyperandrogenemia and age. PRINCIPAL CONCLUSIONS Relying on FPG alone is not adequate for the screening of disorders of glucose tolerance in women with PCOS; such diagnosis should rely on the results of an OGTT regardless of age, weight and/or androgen concentrations.
Collapse
Affiliation(s)
- Andrés E Ortiz-Flores
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, 28034, Spain
| | - Manuel Luque-Ramírez
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, 28034, Spain
| | - Elena Fernández-Durán
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, 28034, Spain
| | - Francisco Alvarez-Blasco
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, 28034, Spain
| | - Héctor F Escobar-Morreale
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, 28034, Spain.
| |
Collapse
|
19
|
Usselman CW, Yarovinsky TO, Steele FE, Leone CA, Taylor HS, Bender JR, Stachenfeld NS. Androgens drive microvascular endothelial dysfunction in women with polycystic ovary syndrome: role of the endothelin B receptor. J Physiol 2019; 597:2853-2865. [PMID: 30847930 DOI: 10.1113/jp277756] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/04/2019] [Indexed: 01/02/2023] Open
Abstract
KEY POINTS Polycystic ovary syndrome (PCOS) is a complex syndrome with cardiovascular risk factors, including obesity and insulin resistance. PCOS is also associated with high androgens, increases the risk of cardiovascular dysfunction in women. Due to the complexity of PCOS, had it has been challenging to isolate specific causes of the cardiovascular dysfunction. Our measure of cardiovascular dysfunction (endothelial dysfunction) was most profound in lean women with PCOS. The endothelin-1-induced vasodilation in these PCOS subject, was dependent on the ETB R but was not NO-dependent. We also demonstrated oestrogen administration improved endothelial function in lean and obese women with PCOS likely because oestrogen increased NO availability. Our studies indicate a primary role for androgens in cardiovascular dysfunction in PCOS. ABSTRACT Endothelin-1 (ET-1) is an indicator of endothelial injury and dysfunction and is elevated in women with androgen excess polycystic ovary syndrome (AE-PCOS). The endothelin B receptor (ETB R) subtype mediates vasodilatation, but is blunted in women with PCOS. We hypothesized that androgen drives endothelial dysfunction in AE-PCOS women and oestradiol (EE) administration reverses these effects. We assessed microvascular endothelial function in women with (7 lean and 7 obese) and without AE-PCOS (controls, 6 lean, 7 obese). Only obese AE-PCOS women were insulin resistant (IR). We evaluated cutaneous vascular conductance (%CVCmax ) with laser Doppler flowmetry during low dose intradermal microdialysis ET-1 perfusions (1, 3, 4, 5 and 7 pmol) with either lactated Ringer solution alone, or with ETB R (BQ-788), or nitric oxide (NO) inhibition (l-NAME). Log[ET-1]-%maxCVC dose-response curves demonstrated reduced vasodilatory responses to ET-1 in lean AE-PCOS (logED50 , 0.59 ± 0.08) versus lean controls (logED50 , 0.49 ± 0.09, P < 0.05), but not compared to obese AE-PCOS (logED50 , 0.65 ± 0.09). ETB R inhibition decreased ET-1-induced vasodilatation in AE-PCOS women (logED50 , 0.64 ± 0. 22, P < 0.05). This was mechanistically observed at the cellular level, with ET-1-induced, DAF-FM-measurable endothelial cell NO production, which was abrogated by dihydrotestosterone in an androgen receptor-dependent manner. EE augmented the cutaneous vasodilating response to ET-1(logED50 0.29 ± 0.21, 0.47 ± 0.09, P < 0.05 for lean and obese, respectively). Androgens drive endothelial dysfunction in lean and obese AE-PCOS. We propose that the attenuated ET-1-induced vasodilatation in AE-PCOS is a consequence of androgen receptor-mediated, suppressed ETB R-stimulated NO production, and is reversed with EE.
Collapse
Affiliation(s)
- Charlotte W Usselman
- John B. Pierce Laboratory, Yale School of Medicine, New Haven, CT, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA.,Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada
| | - Timur O Yarovinsky
- Departments of Internal Medicine (Cardiovascular Medicine) and Immunobiology, Yale School of Medicine, New Haven, CT, USA.,Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Frances E Steele
- Departments of Internal Medicine (Cardiovascular Medicine) and Immunobiology, Yale School of Medicine, New Haven, CT, USA.,Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Cheryl A Leone
- John B. Pierce Laboratory, Yale School of Medicine, New Haven, CT, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Jeffrey R Bender
- Departments of Internal Medicine (Cardiovascular Medicine) and Immunobiology, Yale School of Medicine, New Haven, CT, USA.,Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Nina S Stachenfeld
- John B. Pierce Laboratory, Yale School of Medicine, New Haven, CT, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
20
|
Anik Ilhan G, Yildizhan B, Pekin T. The impact of lipid accumulation product (LAP) and visceral adiposity index (VAI) on clinical, hormonal and metabolic parameters in lean women with polycystic ovary syndrome. Gynecol Endocrinol 2019; 35:233-236. [PMID: 30303693 DOI: 10.1080/09513590.2018.1519794] [Citation(s) in RCA: 8] [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: 01/19/2023] Open
Abstract
This study was performed to assess the impact of lipid accumulation product (LAP) and visceral adiposity index (VAI) on clinical, hormonal, and metabolic parameters in lean women with PCOS. Retrospective analysis of 120 consecutive lean PCOS subjects was performed. Subjects were divided into two groups according to HOMA-IR, as IR + and IR-. A HOMA-IR value above 2.5 was used to indicate IR. Clinical, hormonal and metabolic parameters were compared between the two groups. Correlations between LAP and VAI and clinical, hormonal, metabolic features in women PCOS were analyzed. One hundred twenty lean PCOS subjects were enrolled, of which 39 were insulin resistant. Comparison of group means showed significantly higher values for TG levels, FAI, FGS, TG/HDL-c, TyG, LAP, and VAI indexes and lower values for glucose/insulin ratio and QUICKI in the IR + group. LAP and VAI were both found to be positively correlated with each other and with WC, FAI, FGS, TG, TC levels, lipid ratios, TyG index, and HOMA-IR and negatively correlated with Glucose/Insulin ratio, QUICKI, and HDL-c in lean women with PCOS. LAP and VAI may be promising in early identification of IR and cardiometabolic risk and may be useful for the assessment of hyperandrogenism in lean women with PCOS.
Collapse
Affiliation(s)
- Gokce Anik Ilhan
- a Department of Obstetrics and Gynecology , Marmara University , Istanbul , Turkey
| | - Begum Yildizhan
- a Department of Obstetrics and Gynecology , Marmara University , Istanbul , Turkey
| | - Tanju Pekin
- a Department of Obstetrics and Gynecology , Marmara University , Istanbul , Turkey
| |
Collapse
|
21
|
Speelman DL. Nonpharmacologic Management of Symptoms in Females With Polycystic Ovary Syndrome: A Narrative Review. J Osteopath Med 2019; 119:25-39. [PMID: 30615039 DOI: 10.7556/jaoa.2019.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Polycystic ovary syndrome is the most common hormone disorder in females of reproductive age, affecting reproductive, metabolic, and cardiovascular health. With an unknown cause and a spectrum of common signs and symptoms, diagnosis is based on consensus criteria, and treatment options often target individual symptoms, with variable effectiveness. Safe, effective complementary and alternative therapies can be used to manage symptoms. The first-line intervention is lifestyle modification, including weight loss when appropriate, with caloric restriction and exercise to maintain a healthy weight. Low-carbohydrate and/or low-glycemic index diets can provide additional benefits, and nutritional supplements may be useful adjuncts. The recommended physical activity regimen should include both aerobic and resistance exercise.
Collapse
|
22
|
Cree-Green M, Cai N, Thurston JE, Coe GV, Newnes L, Garcia-Reyes Y, Baumgartner AD, Pyle L, Nadeau KJ. Using simple clinical measures to predict insulin resistance or hyperglycemia in girls with polycystic ovarian syndrome. Pediatr Diabetes 2018; 19:1370-1378. [PMID: 30246333 PMCID: PMC6400639 DOI: 10.1111/pedi.12778] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 08/09/2018] [Accepted: 09/09/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) includes insulin resistance (IR) and impaired glucose tolerance (IGT) in youth, and a greatly elevated risk of type 2 diabetes in adulthood. Identifying IR is challenging and documenting IGT requires an oral glucose tolerance test (OGTT). OBJECTIVE Identify easily applied surrogate measures for IR and IGT in girls with PCOS. METHODS We studied 28 girls with PCOS (body mass index [BMI] percentile 98 (83.99); 15.5 (14.5,16.6) years of age) and 20 with normal menses [BMI percentile (97 (88.99); 15.5 (13.3,16.1) years]. Hyperinsulinemic-euglycemic clamps (insulin dose of 80 μU/ml/min) to determine glucose infusion rate (GIR) and a 75 g OGTT were performed. Surrogates for IR including fasting insulin, homeostatic model assessment-insulin resistant (HOMA-IR), Matsuda index, and estimate of insulin sensitivity (e-IS) were compared to IGT status and GIR. Spearman correlations were performed between surrogates and GIR or IGT, and receiver operator curve (ROC) analysis to predict GIR below the median or IGT status. RESULTS GIR was lower in PCOS (12.9 ± 4.6 vs 17.1 ± 5.1 mg/kg fat-free mass·min; P = 0.01). Within PCOS, HOMA-IR (r = -0.78, P < 0.0001), e-IS (r = 0.70, P < 0.001), and Matsuda (r = 0.533, P < 0.001) correlated with GIR. e-IS provided a good sensitivity (100%) and specificity (71%) to identify IR (e-IS cutoff: <6.3, ROC-area under curve = 0.898). Fasting insulin >22 IU/mL had the best sensitivity (88%), specificity (78%), and ROC (0.760) for IGT status. CONCLUSIONS Girls with PCOS have significant IR, and IGT is common. Both e-IS and fasting insulin are obtainable without an OGTT or clamp and could be used clinically to guide treatment in PCOS.
Collapse
Affiliation(s)
- Melanie Cree-Green
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO.,Center for Women’s Health Research, Aurora, CO
| | - Ninghe Cai
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jessica E. Thurston
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Gregory V. Coe
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Lindsay Newnes
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Yesenia Garcia-Reyes
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Amy D. Baumgartner
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Laura Pyle
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO,Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Kristen J. Nadeau
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO.,Center for Women’s Health Research, Aurora, CO
| |
Collapse
|
23
|
Ibáñez L, Oberfield SE, Witchel S, Auchus RJ, Chang RJ, Codner E, Dabadghao P, Darendeliler F, Elbarbary NS, Gambineri A, Garcia Rudaz C, Hoeger KM, López-Bermejo A, Ong K, Peña AS, Reinehr T, Santoro N, Tena-Sempere M, Tao R, Yildiz BO, Alkhayyat H, Deeb A, Joel D, Horikawa R, de Zegher F, Lee PA. An International Consortium Update: Pathophysiology, Diagnosis, and Treatment of Polycystic Ovarian Syndrome in Adolescence. Horm Res Paediatr 2018; 88:371-395. [PMID: 29156452 DOI: 10.1159/000479371] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/10/2017] [Indexed: 12/11/2022] Open
Abstract
This paper represents an international collaboration of paediatric endocrine and other societies (listed in the Appendix) under the International Consortium of Paediatric Endocrinology (ICPE) aiming to improve worldwide care of adolescent girls with polycystic ovary syndrome (PCOS)1. The manuscript examines pathophysiology and guidelines for the diagnosis and management of PCOS during adolescence. The complex pathophysiology of PCOS involves the interaction of genetic and epigenetic changes, primary ovarian abnormalities, neuroendocrine alterations, and endocrine and metabolic modifiers such as anti-Müllerian hormone, hyperinsulinemia, insulin resistance, adiposity, and adiponectin levels. Appropriate diagnosis of adolescent PCOS should include adequate and careful evaluation of symptoms, such as hirsutism, severe acne, and menstrual irregularities 2 years beyond menarche, and elevated androgen levels. Polycystic ovarian morphology on ultrasound without hyperandrogenism or menstrual irregularities should not be used to diagnose adolescent PCOS. Hyperinsulinemia, insulin resistance, and obesity may be present in adolescents with PCOS, but are not considered to be diagnostic criteria. Treatment of adolescent PCOS should include lifestyle intervention, local therapies, and medications. Insulin sensitizers like metformin and oral contraceptive pills provide short-term benefits on PCOS symptoms. There are limited data on anti-androgens and combined therapies showing additive/synergistic actions for adolescents. Reproductive aspects and transition should be taken into account when managing adolescents.
Collapse
Affiliation(s)
- Lourdes Ibáñez
- Endocrinology, Hospital Sant Joan de Deu, Esplugues, Barcelona, Spain.,CIBERDEM, ISCIII, Madrid, Spain
| | - Sharon E Oberfield
- Division of Pediatric Endocrinology, CUMC, New York-Presbyterian Morgan Stanley Children's Hospital, New York, New York, USA
| | - Selma Witchel
- Division of Pediatric Endocrinology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | | | - R Jeffrey Chang
- Department of Reproductive Medicine, UCSD School of Medicine, La Jolla, California, USA
| | - Ethel Codner
- Institute of Maternal and Child Research, University of Chile, School of Medicine, Santiago, Chile
| | - Preeti Dabadghao
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | | | | | - Alessandra Gambineri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Cecilia Garcia Rudaz
- Division of Women, Youth and Children, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Kathleen M Hoeger
- Department of OBGYN, University of Rochester Medical Center, Rochester, New York, USA
| | - Abel López-Bermejo
- Pediatric Endocrinology, Hospital de Girona Dr. Josep Trueta, Girona, Spain
| | - Ken Ong
- MRC Epidemiology Unit, University of Cambridge, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Alexia S Peña
- The University of Adelaide and Robinson Research Institute, Adelaide, South Australia, Australia
| | - Thomas Reinehr
- University of Witten/Herdecke, Vestische Kinder- und Jugendklinik, Pediatric Endocrinology, Diabetes, and Nutrition Medicine, Datteln, Germany
| | - Nicola Santoro
- Pediatrics, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Rachel Tao
- Division of Pediatric Endocrinology, CUMC, New York-Presbyterian Morgan Stanley Children's Hospital, New York, New York, USA
| | - Bulent O Yildiz
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Hacettepe University School of Medicine, Ankara, Turkey
| | - Haya Alkhayyat
- Medical University of Bahrain, BDF Hospital, Riffa, Bahrein
| | - Asma Deeb
- Mafraq Hospital, Abu Dhabi, United Arab Emirates
| | - Dipesalema Joel
- Department of Paediatrics and Adolescent Health, University of Botswana Teaching Hospital, Gaborone, Botswana
| | - Reiko Horikawa
- Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo, Japan
| | - Francis de Zegher
- Department Pediatrics, University Hospital Gasthuisberg, Leuven, Belgium
| | - Peter A Lee
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
24
|
Gao J, Zhou L, Hong J, Chen C. Assessment of insulin resistance in Chinese PCOS patients with normal glucose tolerance. Gynecol Endocrinol 2017; 33:888-891. [PMID: 28675705 DOI: 10.1080/09513590.2017.1342238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The study aimed to investigate insulin resistance (IR) status in polycystic ovary syndrome (PCOS) women with normal glucose tolerance (NGT), and further to evaluate feasible diagnostic method for those patients. Three hundred and twenty-five PCOS women with NGT and ninety-five healthy age-matched controls were recruited with Rotterdam criterion and 75 g oral glucose tolerance test (OGTT). IR status was estimated following a glycemic and insulinemic OGTT (0, 30, 60, 120, and 180 min). A modified HOMA-IR formula was applied to each time-course value of glycemia and insulinemia. The predictive performance of each IR index was analyzed with the use of ROC curves. Compared with healthy controls, both non-obese and obese PCOS patients with NGT had a higher BMI, serum glucose, insulin value (p < .05). The best predictive index of IR in non-obese PCOS-NGT was a HOMA-M30 value of 20.36 or more (AUC: 0.753). In obese PCOS-NGT population, the best predictive performance was obtained by a HOMA-M60 value of 32.17 or more (AUC: 0.868). IR was common in Chinese PCOS women with NGT, and the early assessment of IR should be heeded. We recommended HOMA-M30 (Cutoff: 20.36) and HOMA-M60 (Cutoff: 32.17) as the best predictive parameters for non-obese and obese PCOS-NGT patients, respectively.
Collapse
Affiliation(s)
- Jing Gao
- a Department of Gynecology and Obstetrics, Rui-Jin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , PR China
- b Department of Gynecology and Obstetrics , The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , PR China
| | - Li Zhou
- a Department of Gynecology and Obstetrics, Rui-Jin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , PR China
| | - Jie Hong
- c Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism , Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , PR China
| | - Chen Chen
- a Department of Gynecology and Obstetrics, Rui-Jin Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , PR China
| |
Collapse
|
25
|
Anastasiou OE, Canbay A, Fuhrer D, Reger-Tan S. Metabolic and androgen profile in underweight women with polycystic ovary syndrome. Arch Gynecol Obstet 2017; 296:363-371. [PMID: 28608050 DOI: 10.1007/s00404-017-4422-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/02/2017] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is a disorder associated with insulin resistance and obesity. Aim of our study is to clarify the prevalence of underweight in PCOS patients and whether metabolic and androgen profiles of PCOS differ depending on normal or low body weight. METHODS Out of 1269 consecutive patients with PCOS recruited from the Department of Endocrinology and Metabolism at the University of Duisburg-Essen, 19 patients (1.5%) were underweight and were compared to 375 lean PCOS subjects (29.6%). Clinical and endocrine parameters were evaluated. Insulin resistance was assessed by 3-h oral glucose tolerance test (OGTT). RESULTS Prevalence of type 2 diabetes and free androgen index did not differ between the two groups. Total cholesterol and low density lipoprotein levels were significantly lower in the group of underweight patients. While no significant difference was found for the Homeostasis model assessment (HOMA) index at fasting state, the HOMA-M120, calculated 2 h after glucose intake in OGTT, was significantly higher in underweight patients. Underweight patients also showed significantly higher postprandial insulin secretion after glucose intake in OGTT. Six underweight PCOS patients received metformin treatment for oligomenorrhea. An improvement of the menstrual cycle was observed in three cases, while two patients were lost to follow up and one discontinued therapy due to side effects. CONCLUSIONS The prevalence of underweight in patients with PCOS is very low. Underweight in PCOS is associated with higher postprandial insulin levels. Several of our underweight patients were able to achieve regular menstrual cycle under metformin therapy.
Collapse
Affiliation(s)
- Olympia E Anastasiou
- Department of Gastroenterology and Hepatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ali Canbay
- Department of Gastroenterology and Hepatology, University Hospital, University of Magdeburg, Essen, Germany
| | - Dagmar Fuhrer
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Susanne Reger-Tan
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
26
|
Song DK, Hong YS, Sung YA, Lee H. Insulin resistance according to β-cell function in women with polycystic ovary syndrome and normal glucose tolerance. PLoS One 2017; 12:e0178120. [PMID: 28542421 PMCID: PMC5444780 DOI: 10.1371/journal.pone.0178120] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/07/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is associated with insulin resistance (IR) and compensatory hyperinsulinemia. IR is recognized as a major risk factor for the development of type 2 diabetes mellitus. However, few studies have investigated IR in women with PCOS and normal glucose tolerance. The objective of this study was to evaluate IR and β-cell function in women with PCOS and normal glucose tolerance. Additionally, we sought to evaluate the usefulness of oral glucose tolerance test (OGTT)-derived IR indices in lean women with PCOS. METHODS We recruited 100 women with PCOS and normal glucose tolerance and 100 age- and BMI-matched women as controls. IR and insulin secretory indices, including the homeostasis-model assessment (HOMA)-IR, HOMA-M120, HOMA-F and the Stumvoll index, were calculated from an OGTT. Increased β-cell function was defined as>75th percentile for the HOMA-F in control women. RESULTS Women with PCOS had higher values for post-load 2-hour glucose, fasting insulin, post-load 2-hour insulin, HOMA-IR, HOMA-M120, HOMA-F and lower values for the Stumvoll index than the controls (all Ps<0.05). Women with PCOS and increased β-cell function showed lower Stumvoll index values than the matched controls (P<0.05). The HOMA-F was significantly associated with the HOMA-M120 and Stumvoll index when adjusted for age and BMI in a multiple regression analysis (all Ps<0.05). The HOMA-M120 was positively correlated with triglycerides and free testosterone, and the Stumvoll index was negatively correlated with triglycerides and free testosterone in lean women with PCOS (all Ps<0.05). CONCLUSIONS Women with PCOS and normal glucose tolerance showed higher IR than controls matched for age, BMI, and β-cell function. β-cell function was increased in women with PCOS when compared to the matched controls, but not when the lean subjects were compared to the matched controls separately. Therefore, early evaluation of IR in women with PCOS and normal glucose tolerance may be needed.
Collapse
Affiliation(s)
- Do Kyeong Song
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul, Korea
| | - Young Sun Hong
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul, Korea
| | - Yeon-Ah Sung
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul, Korea
| | - Hyejin Lee
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
27
|
Goyal M, Dawood AS. Debates Regarding Lean Patients with Polycystic Ovary Syndrome: A Narrative Review. J Hum Reprod Sci 2017; 10:154-161. [PMID: 29142442 PMCID: PMC5672719 DOI: 10.4103/jhrs.jhrs_77_17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex syndrome showing the clinical features of an endocrine/metabolic disorder, including hyperinsulinemia and hyperandrogenism. Two phenotypes are present, either lean or obese, with different biochemical, hormonal, and metabolic profiles. Evidence suggests many treatment modalities that can be applied. However, many of these modalities were found to be not suitable for the lean phenotype of PCOS. Much contradictory research was found regarding lean patients with PCOS. The aim of this narrative review is to shed light on the debate prevailing regarding characteristics, as well as metabolic, hematological, and potential management modalities. Literature review was performed from January 1, 2000 to March 31, 2017 with specific word search such as lean PCOS, hormonal abnormalities in lean PCOS, and the management of lean PCOS. All retrieved articles were carefully assessed, and data were obtained. We could conclude that the debate is still prevailing regarding this specific lean population with PCOS, especially with regard to their characteristics and management modalities. Further studies are still required to resolve this debate on the presence of PCOS in lean women.
Collapse
Affiliation(s)
- Manu Goyal
- Department of Obstetrics and Gynecology, AIIMS, New Delhi, India
| | - Ayman S Dawood
- Department of Obstetrics and Gynecology, Tanta University, Tanta, Egypt
| |
Collapse
|
28
|
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
|
29
|
Greenwood EA, Noel MW, Kao CN, Shinkai K, Pasch LA, Cedars MI, Huddleston HG. Vigorous exercise is associated with superior metabolic profiles in polycystic ovary syndrome independent of total exercise expenditure. Fertil Steril 2015; 105:486-93. [PMID: 26551442 DOI: 10.1016/j.fertnstert.2015.10.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/17/2015] [Accepted: 10/14/2015] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To characterize metabolic features of women with polycystic ovary syndrome (PCOS) by exercise behavior and determine relative health benefits of different exercise intensities. DESIGN Cross-sectional study. SETTING Tertiary academic institution. PATIENT(S) Three hundred and twenty-six women aged 14-52 years-old with PCOS by Rotterdam criteria examined between 2006 and 2013. INTERVENTION(S) International Physical Activity Questionnaire (IPAQ) administered to classify patients into three groups based on Department of Health and Human Services (DHHS) Guidelines of vigorous, moderate, and inactive, along with physical examination and serum testing. MAIN OUTCOME MEASURE(S) Blood pressure, body mass index (BMI), waist circumference, fasting lipids, fasting glucose and insulin, 2-hour 75-gram oral glucose tolerance, homeostatic model assessment of insulin resistance (HOMA-IR). RESULT(S) The DHHS guidelines for adequate physical activity were met by 182 (56%) women. Compared with moderate exercisers and inactive women, the vigorous exercisers had lower BMI and lower HOMA-IR; higher levels of high-density lipoprotein cholesterol and sex hormone-binding globulin; and a reduced prevalence of the metabolic syndrome. In a multivariate logistic regression analysis controlling for age, BMI, and total energy expenditure, every hour of vigorous exercise reduced a patient's odds of metabolic syndrome by 22% (odds ratio 0.78; 95% confidence interval, 0.62, 0.99). CONCLUSION(S) Women with PCOS who met DHHS guidelines for exercise demonstrated superior metabolic health parameters. Vigorous but not moderate activity is associated with reduced odds of the metabolic syndrome, independent of age, BMI, and total energy expenditure. PCOS patients should be encouraged to meet activity guidelines via vigorous physical activity.
Collapse
Affiliation(s)
- Eleni A Greenwood
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California at San Francisco, San Francisco, California.
| | - Martha W Noel
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California at San Francisco, San Francisco, California
| | - Chia-Ning Kao
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California at San Francisco, San Francisco, California
| | - Kanade Shinkai
- Department of Dermatology, University of California at San Francisco, San Francisco, California
| | - Lauri A Pasch
- Department of Psychiatry, University of California at San Francisco, San Francisco, California
| | - Marcelle I Cedars
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California at San Francisco, San Francisco, California
| | - Heather G Huddleston
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California at San Francisco, San Francisco, California
| |
Collapse
|
30
|
Liu W, Liu W, Fu Y, Wang Y, Zhang Y. Bak Foong pills combined with metformin in the treatment of a polycystic ovarian syndrome rat model. Oncol Lett 2015; 10:1819-1825. [PMID: 26622758 DOI: 10.3892/ol.2015.3466] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 04/14/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the treatment effects and associated mechanism of Bak Foong pills (BFPs) combined with metformin in the treatment of polycystic ovarian syndrome (PCOS). BFPs and/or metformin were administrated to treat the PCOS rats, and the weights and morphologies of the ovary, uterus and adrenal gland were measured. The levels of fasting blood glucose (FBG), serum testosterone (T), luteinizing hormone, fasting insulin (FIN) and insulin-like growth factor-1 were also measured, and the homeostasis model assessment of insulin resistance (HOMA-IR) was calculated. The expression level of androgen receptor (AR) in the ovarian tissue, and the cytochrome P450 cholesterol side-chain cleavage enzyme (P450scc) mRNA levels in the ovary and adrenal tissues were detected. The levels of T, FIN, FBG and HOMA-IR in the combination group were significantly reduced; the wet weights of the ovary and the adrenal gland were decreased significantly, while that of the uterus was increased, and the histological morphology benignly recovered. The rats of each treatment group all experienced restored ovulation. The AR expression level in the treatment group was reduced, and the P450scc mRNA levels in the ovary and the adrenal gland of the combined treatment group were decreased. BFPs combined with metformin significantly affected PCOS, and the possible mechanism involved in the treatment may have been through the reduction of P450scc generation. BFPs may reduce the androgen levels, thus allowing the ovary to restore ovulation.
Collapse
Affiliation(s)
- Wenhui Liu
- Center of Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wenpei Liu
- Translational Medicine Institute, Affiliated to The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan 423000, P.R. China
| | - Yuling Fu
- Department of Obstetrics and Gynecology, The Second Hospital of Yinzhou, Ningbo, Zhejiang 315000, P.R. China
| | - Yan Wang
- Center of Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yuanzhen Zhang
- Center of Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|