51
|
Scott D, Harrison CL, Hutchison S, de Courten B, Stepto NK. Exploring factors related to changes in body composition, insulin sensitivity and aerobic capacity in response to a 12-week exercise intervention in overweight and obese women with and without polycystic ovary syndrome. PLoS One 2017; 12:e0182412. [PMID: 28771628 PMCID: PMC5542389 DOI: 10.1371/journal.pone.0182412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/07/2017] [Indexed: 01/04/2023] Open
Abstract
Objective To determine factors associated with differential changes in body fat, insulin resistance and aerobic capacity following a 12-week exercise intervention in overweight and obese women with and without polycystic ovary syndrome (PCOS). Methods 16 overweight and obese women (9 PCOS; 7 without PCOS) completed a supervised progressive 12-week exercise program. Primary outcomes included changes in indicators of insulin sensitivity (including glucose infusion rate relative to fat-free mass [GIR/FFM]), body composition, and aerobic capacity (VO2 peak; 12 participants only). Comparisons were made between women with and without PCOS, and between participants who lost ≥5% (classified as exercise responders) and <5% (non-responders) in body fat (assessed by dual-energy X-ray absorptiometry). Results Training decreased body fat percentage by (mean; 95% CI) -2.3%; -5.3, 0.7% in women with PCOS and by -6.4%; -10.9, -1.9% in women without PCOS (P = 0.08). Ten women (7 PCOS; 3 without PCOS) did not reduce body fat by ≥5%. All participants improved VO2 peak (mean change 27%; 16–39%) but four (2 PCOS; 2 without PCOS) demonstrated decreases in GIR/FFM (mean change for whole cohort: 37%; 3–71%). Android-gynoid fat ratio (0.58; 0.51, 0.66 vs 0.46; 0.40, 0.51; P<0.01) was significantly higher and GIR/FFM (6.69; 3.49, 9.90 vs 11.44; 9.15, 13.72 mg/kg/min; P = 0.01) was significantly lower in non-responders compared with responders at baseline, but non-responders had significant post-training decreases in android-gynoid ratio (-0.02; -0.04, -0.01; P = 0.03), and increases in VO2 peak (7.24; 2.28, 12.21 mL/kg/min; P = 0.01) and GIR/FFM (1.44; 0.27, 2.61 mg/kg/min; P = 0.02). In women with PCOS, pre-training VO2 peak was significantly negatively correlated with change in total body fat (r = -0.75; P = 0.02), and pre-training fasting glucose negatively correlated with changes in VO2 peak (r = -0.76; P = 0.04), but positively correlated with changes in GIR (r = 0.67; P = 0.046). Conclusion A high proportion of overweight and obese women with PCOS had small reductions in body fat following a 12-week exercise intervention, but nevertheless significantly reduced relative central adiposity and improved aerobic capacity and insulin sensitivity.
Collapse
Affiliation(s)
- David Scott
- Department of Medicine, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
- Department of Medicine – Western Campus and Australian Institute for Musculoskeletal Science, The University of Melbourne, St Albans, Victoria, Australia
- * E-mail:
| | - Cheryce L. Harrison
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, Victoria, Australia
| | - Samantha Hutchison
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, Victoria, Australia
- Diabetes and Vascular Medicine Unit, Monash Health, Clayton, Victoria, Australia
| | - Barbora de Courten
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, Victoria, Australia
- Diabetes and Vascular Medicine Unit, Monash Health, Clayton, Victoria, Australia
| | - Nigel K. Stepto
- Department of Medicine – Western Campus and Australian Institute for Musculoskeletal Science, The University of Melbourne, St Albans, Victoria, Australia
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, Victoria, Australia
- Institute of Sport Exercise and Active Living (ISEAL) Victoria University, Melbourne, Victoria, Australia
| |
Collapse
|
52
|
Zheng X, Chen J, Xie T, Xia Z, Loo WTY, Lao L, You J, Yang J, Tsui K, Mo F, Gao F. Relationship between Chinese medicine dietary patterns and the incidence of breast cancer in Chinese women in Hong Kong: a retrospective cross-sectional survey. Chin Med 2017; 12:17. [PMID: 28670332 PMCID: PMC5492296 DOI: 10.1186/s13020-017-0138-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/06/2017] [Indexed: 01/20/2023] Open
Abstract
Background This retrospective cross-sectional study aimed to investigate the relationship between Chinese medicine (CM) dietary patterns (hot, neutral, and cold) and the incidence of breast cancer among Chinese women in Hong Kong. Methods Breast cancer cases (n = 202) and healthy controls (n = 202) were matched according to demographics. Chinese women residing in Hong Kong for the past 7 years were recruited by media advertisements (e.g., via newspapers, radio, and posters). The control participants were recruited by convenience sampling from health workshops held in clinics and communities of 15 districts of Hong Kong. After completing test–retest reliability, all participants were asked to complete diet pattern questionnaires about their food preferences and dietary patterns. The Student’s unpaired t test, Chi square test, and logistic regression were conducted using SPSS software. Results Three major CM dietary patterns were identified: hot, neutral, and cold. The participants with breast cancer exhibited a stronger preference for hot food than the control group (Chi square test, P < 0.001). A higher frequency of breast cancer was associated with a higher frequency of dining out for breakfast (4–5 times per week, Chi square test, P = 0.015; 6–7 times per week, Chi square test, P < 0.001) and lunch (4–5 times per week, Chi square test, P < 0.001; 6–7 times per week, Chi square test, P = 0.006). The participants with no history of breast cancer consumed CM supplements and Guangdong soups (1–2 times per week, Chi square test, P = 0.05; >3 times per week, Chi square test, P < 0.001) more frequently than those with breast cancer. Conclusions Non-breast cancer participants adopted a neutral (healthy and balanced) dietary pattern, and consumed CM supplements and Guangdong soups more frequently. Electronic supplementary material The online version of this article (doi:10.1186/s13020-017-0138-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiao Zheng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.,Department of Dermatology, Guangzhou University of Chinese Medicine, Guangzhou, 510020 China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.,Chengdu University of Traditional Chinese Medicine, Chengdu, 510020 China
| | - Ting Xie
- Department of Dermatology, Guangzhou University of Chinese Medicine, Guangzhou, 510020 China
| | - Zhiyu Xia
- School of Public Health, Peking University, Beijing, China
| | | | - Lixing Lao
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - JieShu You
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Jie Yang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 510020 China
| | - Kamchuen Tsui
- The Hong Kong Associate of Chinese Medicine, Hong Kong, China
| | - Feizhi Mo
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Fei Gao
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
53
|
Successful treatment with Korean herbal medicine and lifestyle management in an obese woman with polycystic ovarian syndrome. Integr Med Res 2017; 6:325-328. [PMID: 28951847 PMCID: PMC5605367 DOI: 10.1016/j.imr.2017.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/05/2017] [Accepted: 06/15/2017] [Indexed: 11/27/2022] Open
Abstract
In Korea, herbal remedies have been widely used to treat polycystic ovarian syndrome (PCOS). We report the case of a woman with obese-type PCOS who was successfully treated with Korean herbal medicine (KHM) and lifestyle management. A 23-year-old female patient was first examined at our clinic in April 2015. She had amenorrhea for the previous 9 months and presented with obesity and abnormal reproductive hormone levels. She was treated using KHM and lifestyle management. We observed her menstrual cycles and re-evaluated her hormonal levels during the treatment. After KHM therapy, her body weight decreased from 88.2 kg to 66.7 kg. Her menstrual cycles resumed regularly, and the serum levels of the hormones had normalized. No adverse effects on liver and renal functions were observed. This study indicates that KHM might be considered an option for treating women with obese-type PCOS. Further large-scale trials are needed.
Collapse
|
54
|
Reinehr T, Kulle A, Rothermel J, Knop-Schmenn C, Lass N, Bosse C, Holterhus PM. Longitudinal analyses of the steroid metabolome in obese PCOS girls with weight loss. Endocr Connect 2017; 6:213-224. [PMID: 28373267 PMCID: PMC5435851 DOI: 10.1530/ec-17-0051] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 04/03/2017] [Indexed: 01/01/2023]
Abstract
OBJECTIVE The underlying mechanisms of polycystic ovarian syndrome (PCOS) are not fully understood yet. The aim of the study was to get functional insights into the regulation of steroid hormones in PCOS by steroid metabolomics. DESIGN This is a longitudinal study of changes of steroid hormones in 40 obese girls aged 13-16 years (50% with PCOS) participating in a 1-year lifestyle intervention. Girls with and without PCOS were matched to age, BMI and change of weight status. METHODS We measured progesterone, 17-hydroxyprogesterone, 17-hydroxyprogenolon, 11-deoxycorticosterone, 21-deoxycorticosterone, deoxycorticosterone, corticosterone, 11-deoxycortisol, cortisol, cortisone, androstenedione, testosterone, dehydroepiandrostendione-sulfate (DHEA-S), estrone and estradiol by LC-MS/MS steroid profiling at baseline and one year later. RESULTS At baseline, obese PCOS girls demonstrated significantly higher androstenedione and testosterone concentrations compared to obese girls without PCOS, whereas the other steroid hormones including glucocorticoids, mineralocorticoids, estrogens and precursors of androgens did not differ significantly. Weight loss in obese PCOS girls was associated with a significant decrease of testosterone, androstenedione, DHEA-S, cortisol and corticosterone concentrations. Weight loss in obese non-PCOS girls was associated with a significant decrease of DHEA-S, cortisol and corticosterone concentrations, whereas no significant changes of testosterone and androstenedione concentrations could be observed. Without weight loss, no significant changes of steroid hormones were measured except an increase of estradiol in obese PCOS girls without weight loss. CONCLUSIONS The key steroid hormones in obese adolescents with PCOS are androstenedione and testosterone, whereas glucocorticoids, mineralocorticoids, estrogens and precursors of androgens did not differ between obese girls with and without PCOS.
Collapse
Affiliation(s)
- Thomas Reinehr
- Department of Pediatric EndocrinologyDiabetes, and Nutrition Medicine, Vestische Hospital for Children and Adolescents, University of Witten/Herdecke, Datteln, Germany
| | - Alexandra Kulle
- Division of Pediatric Endocrinology and DiabetesDepartment of Pediatrics, University Hospital of Schleswig - Holstein, UKSH, Campus Kiel/Christian Albrechts University of Kiel, CAU, Kiel, Germany
| | - Juliane Rothermel
- Department of Pediatric EndocrinologyDiabetes, and Nutrition Medicine, Vestische Hospital for Children and Adolescents, University of Witten/Herdecke, Datteln, Germany
| | - Caroline Knop-Schmenn
- Department of Pediatric EndocrinologyDiabetes, and Nutrition Medicine, Vestische Hospital for Children and Adolescents, University of Witten/Herdecke, Datteln, Germany
| | - Nina Lass
- Department of Pediatric EndocrinologyDiabetes, and Nutrition Medicine, Vestische Hospital for Children and Adolescents, University of Witten/Herdecke, Datteln, Germany
| | - Christina Bosse
- Department of Pediatric EndocrinologyDiabetes, and Nutrition Medicine, Vestische Hospital for Children and Adolescents, University of Witten/Herdecke, Datteln, Germany
| | - Paul-Martin Holterhus
- Division of Pediatric Endocrinology and DiabetesDepartment of Pediatrics, University Hospital of Schleswig - Holstein, UKSH, Campus Kiel/Christian Albrechts University of Kiel, CAU, Kiel, Germany
| |
Collapse
|
55
|
Dokras A, Playford M, Kris-Etherton PM, Kunselman AR, Stetter CM, Williams NI, Gnatuk CL, Estes SJ, Sarwer DB, Allison KC, Coutifaris C, Mehta N, Legro RS. Impact of hormonal contraception and weight loss on high-density lipoprotein cholesterol efflux and lipoprotein particles in women with polycystic ovary syndrome. Clin Endocrinol (Oxf) 2017; 86:739-746. [PMID: 28199736 PMCID: PMC5651984 DOI: 10.1111/cen.13310] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/04/2017] [Accepted: 02/10/2017] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To study the effects of oral contraceptive pills (OCP), the first-line treatment for PCOS, on high-density lipoprotein cholesterol (HDL-C) function (reverse cholesterol efflux capacity) and lipoprotein particles measured using nuclear magnetic resonance spectroscopy in obese women. DESIGN Secondary analysis of a randomized controlled trial (OWL-PCOS) of OCP or Lifestyle (intensive Lifestyle modification) or Combined (OCP + Lifestyle) treatment groups for 16 weeks. PATIENTS Eighty-seven overweight/obese women with PCOS at two academic centres. MEASUREMENTS Change in HDL-C efflux capacity and lipoprotein particles. RESULTS High-density lipoprotein cholesterol efflux capacity increased significantly at 16 weeks in the OCP group [0·11; 95% confidence interval (CI) 0·03, 0·18, P = 0·008] but not in the Lifestyle (P = 0·39) or Combined group (P = 0·18). After adjusting for HDL-C and TG levels, there was significant mean change in efflux in the Combined group (0·09; 95% CI 0·01, 0·15; P = 0·01). Change in HDL-C efflux correlated inversely with change in serum testosterone (rs = -0·21; P = 0·05). In contrast, OCP use induced an atherogenic low-density lipoprotein cholesterol (LDL-C) profile with increase in small (P = 0·006) and large LDL-particles (P = 0·002). Change in small LDL-particles correlated with change in serum testosterone (rs = -0·31, P = 0·009) and insulin sensitivity index (ISI; rs = -0·31, P = 0·02). Both Lifestyle and Combined groups did not show significant changes in the atherogenic LDL particles. CONCLUSIONS Oral contraceptive pills use is associated with improved HDL-C function and a concomitant atherogenic LDL-C profile. Combination of a Lifestyle program with OCP use improved HDL-C function and mitigated adverse effects of OCP on lipoproteins. Our study provides evidence for use of OCP in overweight/obese women with PCOS when combined with Lifestyle changes.
Collapse
Affiliation(s)
- Anuja Dokras
- The Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Martin Playford
- Section of Inflammation and Cardiometabolic Disease, National Heart, Lung and Blood Institute, Bethesda, MA, USA
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, Penn State College of Health and Human Development, University Park, Philadelphia, PA, USA
| | - Allen R Kunselman
- Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Christy M Stetter
- Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Nancy I Williams
- Department of Kinesiology, Penn State College of Health and Human Development, University Park, Philadelphia, PA, USA
| | - Carol L Gnatuk
- Department of Obstetrics and Gynecology, Penn State College of Medicine, Hershey, PA, USA
| | - Stephanie J Estes
- Department of Obstetrics and Gynecology, Penn State College of Medicine, Hershey, PA, USA
| | - David B Sarwer
- Department of Psychiatry, Temple University, Philadelphia, PA, USA
- Department of Social and Behavioral Science, Center for Obesity Research and Education, College of Public Health, Temple University, Philadelphia, PA, USA
| | - Kelly C Allison
- Department of Psychiatry, Temple University, Philadelphia, PA, USA
| | - Christos Coutifaris
- The Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Nehal Mehta
- Section of Inflammation and Cardiometabolic Disease, National Heart, Lung and Blood Institute, Bethesda, MA, USA
| | - Richard S Legro
- Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
- Department of Obstetrics and Gynecology, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
56
|
Jiskoot G, Benneheij SH, Beerthuizen A, de Niet JE, de Klerk C, Timman R, Busschbach JJ, Laven JSE. A three-component cognitive behavioural lifestyle program for preconceptional weight-loss in women with polycystic ovary syndrome (PCOS): a protocol for a randomized controlled trial. Reprod Health 2017; 14:34. [PMID: 28264692 PMCID: PMC5339998 DOI: 10.1186/s12978-017-0295-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 02/22/2017] [Indexed: 12/24/2022] Open
Abstract
Background Obesity in women with polycystic ovary syndrome (PCOS) negatively affects all clinical features, and a 5 to 10% weight loss has shown promising results on reproductive, metabolic and psychological level. Incorporating a healthy diet, increasing physical activity and changing dysfunctional thought patterns in women with PCOS are key points in losing weight. The biggest challenge in weight management programs is to achieve a reasonable and sustainable weight loss. The aim of this study is to explore whether Cognitive Behavioural Therapy (CBT) by a mental health professional, working in a multidisciplinary team with a dietician and a physical therapist (a three-component intervention), is more effective for weight loss in the long term, within 12 months. We will also explore whether mobile phone applications are effective in supporting behavioural change and sustainable weight loss. Methods The present study is a longitudinal randomized controlled trial (RCT) to study the effectiveness of a three-component 1-year cognitive-behavioural lifestyle intervention in overweight/obese women with PCOS. A total of 210 participants are randomly assigned to three groups: 1) CBT provided by the multidisciplinary team or; 2) CBT provided by the multidisciplinary team and Short Message Service (SMS) or; 3) usual care: encourage weight loss through publicly available services (control group). The primary aim of the 12-month intervention is to explore whether a three-component 1-year cognitive-behavioural lifestyle intervention is effective to decrease weight, when compared to usual care. Secondary outcomes include: the effect of the intervention on the PCOS phenotype, waist circumference, waist to hip ratio, ovulation rates, total testosterone, SHBG, free androgen index (FAI), AMH, hirsutism, acne, fasting glucose, blood pressure and all psychological parameters. Additionally, we assessed time to pregnancy, ongoing pregnancies, clinical pregnancies, miscarriages and birth weight. All outcome variables are measured at the start of the study, and again at 3 months, 6 months, nine months and 12 months. Discussion We expect that CBT provided by a multidisciplinary team, especially combined with SMS, is effective in developing a healthy lifestyle and achieving a long-term weight loss in women with PCOS. Losing 5– 10% body weight improves various PCOS characteristics. Consequently, we expect to show that CBT provided by a multidisciplinary team improves reproductive and metabolic outcomes, as well as quality of life, while at the same time being cost-effective. Trial registration Registered at the Netherlands National Trial Register with number NTR2450 on August 2nd, 2010.
Collapse
Affiliation(s)
- G Jiskoot
- Division Reproductive Medicine, Department of Obstetrics and Gynaecology, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands. .,Department of Psychiatry, Section of Medical Psychology and Psychotherapy, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands.
| | - S H Benneheij
- Division Reproductive Medicine, Department of Obstetrics and Gynaecology, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - A Beerthuizen
- Department of Psychiatry, Section of Medical Psychology and Psychotherapy, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - J E de Niet
- Division Reproductive Medicine, Department of Obstetrics and Gynaecology, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands.,Department of Psychiatry, Section of Medical Psychology and Psychotherapy, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - C de Klerk
- Department of Psychiatry, Section of Medical Psychology and Psychotherapy, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - R Timman
- Department of Psychiatry, Section of Medical Psychology and Psychotherapy, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - J J Busschbach
- Department of Psychiatry, Section of Medical Psychology and Psychotherapy, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - J S E Laven
- Division Reproductive Medicine, Department of Obstetrics and Gynaecology, Erasmus MC, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| |
Collapse
|
57
|
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: 33] [Impact Index Per Article: 4.1] [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
|
58
|
Mirabolghasemi G, Kamyab Z. Changes of The Uterine Tissue in Rats with Polycystic Ovary Syndrome Induced by Estradiol Valerate. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2016; 11:47-55. [PMID: 28367305 PMCID: PMC5215711 DOI: 10.22074/ijfs.2016.4794] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 07/26/2016] [Indexed: 11/04/2022]
Abstract
Background Polycystic ovary syndrome (PCOS) is one of the most common hormonal
disorders that can lead to irregular menstrual cycles and hyperandrogenism. Reduced
levels of progesterone and increased estrogen in these women can perpetually stimulate
the endometrial tissue of the uterus. In this study, we assess the effect of PCOS induction
by estradiol valerate (EV) in a rat model. Materials and Methods In this experimental study, adult female Wistar rats that weighed
approximately 200 g were divided into control, sham, and experimental groups (n=6 per
group). The experimental group received subcutaneous injections of 2 mg EV for induction
of PCOS. We confirmed the presence of PCOS in the experimental group rats. Rats from all
groups were subsequently killed, after which their uteri were removed and fixed for histological and cytological analyses. The uterine tissue sections were stained with hematoxylin
and eosin (H&E) and iron hematoxylin (iron-H). We examined epithelium height, thickness
of the uterus wall, and frequency of the mitotic cells. The data were assessed at α=0.05. Results Uterine tissue findings from the experimental group showed significant increases
in the height of the uterus luminal epithelium, the thickness of the uterus wall, and the frequency of eosinophils in the endometrial stroma. We observed an increased frequency of
mitotic cells in the experimental group in both luminal and glandular epithelia of the uterus.
An increased rate of the glandular epithelium region was noticeable and significant. Conclusion Induction of PCOS by EV could change the proliferation rate in the endo-
metrial tissue of the uterus.
Collapse
Affiliation(s)
- Ghadire Mirabolghasemi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Zahra Kamyab
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| |
Collapse
|
59
|
Roh EY, Yoon JH, Song EY, Kim JJ, Hwang KR, Seo SH, Shin S. Single nucleotide polymorphisms in the TGF-β1 gene are associated with polycystic ovary syndrome susceptibility and characteristics: a study in Korean women. J Assist Reprod Genet 2016; 34:139-147. [PMID: 27796807 DOI: 10.1007/s10815-016-0824-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/30/2016] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Although many hypotheses regarding the pathogenesis of polycystic ovary syndrome (PCOS) have been generated, genetic studies have not identified specific genes that play a role in PCOS etiopathogenesis. This study aimed to investigate the relationship between TGF-β1 gene polymorphism and PCOS in Koreans. METHOD A total of 51 Korean women with PCOS and 69 healthy women were enrolled. We analyzed 4 single nucleotide polymorphisms (SNPs) of the TGF-β1 gene (rs11466313, rs1800469, rs2317130, and rs4803457). We also analyzed laboratory measurements, such as free testosterone, glucose, and cholesterol. RESULTS The frequencies of rs1800469T allele negativity, rs4803457T allele negativity, the rs1800469CC genotype, and the rs4803457CC genotype showed positive associations with PCOS (P = 0.003, P = 0.027, P = 0.009, and P=0.031, respectively), whereas the haplotypes rs1800469C-rs4803457T and rs1800469T-rs4803457T showed negative associations with PCOS. A strong protective effect of the "rs1800469CT-rs4803457TT" combination (OR = 0.09) and a strong risk effect of "rs1800469CC-rs4803457CC" (OR = 6.23) for PCOS were observed. The rs1800469C/T and rs2317130C/T SNPs exhibited associations with several laboratory measurements with various levels of significance. CONCLUSION The results demonstrated an association of TGF-β1 gene polymorphisms with the development and/or characteristics of PCOS in the Korean population.
Collapse
Affiliation(s)
- Eun Youn Roh
- Department of Laboratory Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea.,Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong Hyun Yoon
- Department of Laboratory Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea.,Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun Young Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin Ju Kim
- Department of Obstetrics and Gynecology, Seoul National University Healthcare System Gangnam Center, Seoul, Republic of Korea
| | - Kyu Ri Hwang
- Departments of Obstetrics and Gynecology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Soo Hyun Seo
- Department of Laboratory Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea.,Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sue Shin
- Department of Laboratory Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea. .,Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
60
|
Xiang Y, Song Y, Li Y, Zhao D, Ma L, Tan L. miR-483 is Down-Regulated in Polycystic Ovarian Syndrome and Inhibits KGN Cell Proliferation via Targeting Insulin-Like Growth Factor 1 (IGF1). Med Sci Monit 2016; 22:3383-3393. [PMID: 27662007 PMCID: PMC5040236 DOI: 10.12659/msm.897301] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Polycystic ovarian syndrome (PCOS) is a common metabolic disorder in premenopausal woman, characterized by hyperandrogenism, oligoanovulation, and insulin resistance. microRNAs play pivotal roles in regulating key factors of PCOS. However, relevant research remains limited. This study aimed to reveal the role and potential mechanism of miR-483 in PCOS. Material/Methods PCOS patients (n=20) were recruited for detecting miR-483 expression in lesion and normal ovary cortex. Human granulosa-like tumor cell line KGN was used to alter miR-483 expression by cell transfection. Cell viability and proliferation were analyzed by MTT assay and colony formation assay, and cell cycle was detected by flow cytometry. Interaction between miR-483 and IGF1 was verified by luciferase reporter assay. KGN cells were further treated by insulin to investigate the relationship between miR-483 and insulin. Results miR-483 was significantly down-regulated in lesion ovary cortex from PCOS patients (P<0.001). In KGN cells, overexpression of miR-483 inhibited cell viability and proliferation, and induced cell cycle arrest. miR-483 also inhibited CCNB1, CCND1, and CDK2. miR-483 sponge induced the opposite effects. miR-483 directly targeted IGF1 3′UTR, and IGF1 promoted KGN cell proliferation and reversed miR-483-inhibited cell viability. Insulin treatment in KGN cells inhibited miR-483, and promoted IGF1 and cell proliferation. Conclusions These results suggest that miR-483 is a PCOS suppressor inhibiting cell proliferation, possibly via targeting IGF1, and that it is involved in insulin-induced cell proliferation. miR-483 is a potential alternative for diagnosing and treating PCOS.
Collapse
Affiliation(s)
- Yungai Xiang
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Yuxia Song
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Yan Li
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Dongmei Zhao
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Liying Ma
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Li Tan
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| |
Collapse
|
61
|
Shishehgar F, Ramezani Tehrani F, Mirmiran P, Hajian S, Baghestani AR, Moslehi N. Comparison of Dietary Intake between Polycystic Ovary Syndrome Women and Controls. Glob J Health Sci 2016; 8:54801. [PMID: 27157182 PMCID: PMC5064084 DOI: 10.5539/gjhs.v8n9p302] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 12/28/2015] [Accepted: 12/14/2015] [Indexed: 12/24/2022] Open
Abstract
Polycystic Ovary syndrome (PCOS) is a complicated endocrinopathy affecting women in reproductive age. The crucial role of obesity and insulin resistance in progression of metabolic and cardiovascular features of PCOS has been confirmed. Although it has been suggested that there is a possible association between dietary pattern and risk of PCOS, few studies investigating the diet composition of PCOS women. The aim of this study was to compare the dietary intakes between women with polycystic ovary syndrome (PCOS) and eumenorrheic non hirsute women. This was a case control study of 142 women with PCOS and 140 eumenorrheic non hirsute healthy age and BMI matched controls. We compared the dietary intakes of our study group using a validated food frequency questionnaire (FFQ), using T-test or Mann-Whitney to compare the means of two groups. One way Anova was used to compare the tertiles of GI and GL in each group and a two way ANOVA was used to compare between tertiles of GI-GL and groups. The results demonstrated that energy and macronutrient intakes in PCOS women compared to controls were similar. PCOS group consumed more food items with high glycemic index (p=0.042) and less legumes (P=0.026) and vegetables (p=0.037) than controls. Both groups in the highest tertile of glycemic load (GL) had higher body mass index and waist circumference. Considering the results of this study, it was concluded that PCOS women had a dietary pattern that was characterized by a higher consumption of high GI food items and lower legumes and vegetables.
Collapse
|
62
|
de Barros Machado A, Dos Reis V, Weber S, Jauckus J, Brum IS, von Eye Corleta H, Strowitzki T, Capp E, Germeyer A. Proliferation and metastatic potential of endometrial cancer cells in response to metformin treatment in a high versus normal glucose environment. Oncol Lett 2016; 12:3626-3632. [PMID: 27900046 DOI: 10.3892/ol.2016.5041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/22/2016] [Indexed: 11/06/2022] Open
Abstract
In order to improve our understanding of the potential preventive and therapeutic role of metformin, the present study aimed to investigate the capability of low-dose metformin in the efficient inhibition of cancer development and the reduction of the metastasis of endometrial adenocarcinoma type I and primary endometrial epithelial cells (eEPs), with the drug acting as a treatment in a hyperinsulinemic environment exposed to high and normal glucose conditions. The Ishikawa endometrial adenocarcinoma cell line and primary eEPs were exposed to an environment with high (17 mM) or normal glucose (5 mM) and treated with insulin, low-dose metformin (0.1 mM) or a combined treatment. Metastatic potential was assessed by migration and invasion assays, and relative cell proliferation was determined. Metformin at a low dose potently inhibited the insulin action, decreasing the ability of the endometrial cancer (EC) cell line to migrate and invade in a high and normal glucose environment, and decreasing the migration ability of the primary eEPs. In the EC cell line, the insulin treatment increased the proliferation, without any subsequent reduction of proliferation by the addition of 0.1 mM metformin; however, relative cell proliferation sensitivity to metformin was observed in the range between 1 and 5 mM regardless of the glucose concentration present. Overall, metformin at 0.1 mM is not efficient enough to decrease the proliferation in an EC cell line. However, at this concentration, metformin can inhibit the insulin action in endometrial epithelial cancer cells, demonstrating an anti-metastatic effect in high and normal glucose environments.
Collapse
Affiliation(s)
- Amanda de Barros Machado
- Laboratory of Molecular Endocrine Biology and Tumor Biology, Department of Physiology, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Vania Dos Reis
- Laboratory of Molecular Endocrine Biology and Tumor Biology, Department of Physiology, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Sebastian Weber
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Julia Jauckus
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Ilma Simoni Brum
- Laboratory of Molecular Endocrine Biology and Tumor Biology, Department of Physiology, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Helena von Eye Corleta
- Laboratory of Molecular Endocrine Biology and Tumor Biology, Department of Physiology, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Thomas Strowitzki
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Edison Capp
- Laboratory of Molecular Endocrine Biology and Tumor Biology, Department of Physiology, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Ariane Germeyer
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| |
Collapse
|
63
|
Effect of meal frequency on glucose and insulin levels in women with polycystic ovary syndrome: a randomised trial. Eur J Clin Nutr 2016; 70:588-94. [PMID: 26862008 DOI: 10.1038/ejcn.2015.225] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 11/04/2015] [Accepted: 11/20/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND/OBJECTIVES The aim of the study was to compare the effect of two-meal patterns (three vs six meals per day) on glucose and insulin levels in women with polycystic ovary syndrome (PCOS). SUBJECTS/METHODS In a randomised, crossover, 24-week study, 40 women with PCOS, aged 27±6 years, body mass index 27±6 kg/m(2), followed a weight maintenance diet (% carbohydrates:protein:fat, 40:25:35), consumed either as a three- or a six-meal pattern, with each intervention lasting for 12 weeks. Anthropometric measurements, diet compliance and subjective hunger, satiety and desire to eat were assessed biweekly. All women underwent an oral glucose tolerance test (OGTT) with 75 g glucose for measurement of plasma glucose and insulin at the beginning and end of each intervention. HaemoglobinA1c (HbA1c), blood lipids and hepatic enzymes were measured at the beginning and end of each intervention. RESULTS Body weight remained stable throughout the study. Six meals decreased significantly fasting insulin (P=0.014) and post-OGTT insulin sensitivity (Matsuda index, P=0.039) vs three meals. After incorporation of individual changes over time, with adjustment for potential confounders, the only variable that remained significant was the Matsuda index, which was then used in multivariate analysis and general linear models. Six meals improved post-OGTT insulin sensitivity independently of age and body weight vs three meals (P=0.012). No significant differences were found between six and three meals for glucose, HbA1c, blood lipids, hepatic enzymes, subjective desire to eat and satiety. CONCLUSIONS Six meals had a more favourable effect on post-OGTT insulin sensitivity in women with PCOS compared with isocaloric three meals.
Collapse
|
64
|
Li X, Pishdari B, Cui P, Hu M, Yang HP, Guo YR, Jiang HY, Feng Y, Billig H, Shao R. Regulation of Androgen Receptor Expression Alters AMPK Phosphorylation in the Endometrium: In Vivo and In Vitro Studies in Women with Polycystic Ovary Syndrome. Int J Biol Sci 2015; 11:1376-89. [PMID: 26681917 PMCID: PMC4671995 DOI: 10.7150/ijbs.13109] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/12/2015] [Indexed: 11/05/2022] Open
Abstract
The failure of reproductive success in polycystic ovary syndrome (PCOS) patients could be in part due to endometrial dysfunction. However, no studies have investigated any causality between androgen, androgen receptor (AR) expression, and adenosine monophosphate activated protein kinase (AMPK) activation in the endometrium under physiological and pathological conditions. In the present study, we show that 1) endometrial AR expression levels fluctuate in non-PCOS and PCOS patients during the menstrual cycle; 2) the menstrual phase-dependent alteration of p-AMPKα expression occurs in non-PCOS patients but not in PCOS patients; 3) AR expression is higher in PCOS patients than non-PCOS patients during hyperplasia while AMPKα activation (indicated by the ratio of p-AMPKα to AMPKα); and 4) co-localization of AR and Ki-67 in epithelial cell nuclei is observed in endometrial hyperplasia. Importantly, using in vitro human tissue culture and an in vivo 5α-dihydrotestosterone-treated rat model, we show that the action of androgen on AMPKα activation is likely mediated through nuclear AR, especially in epithelial cells. Collectively, we present evidence that AR expression and AMPKα activation depend on menstrual cycle phase and the presence of PCOS, and the data suggest that AR-mediated regulation of AMPKα activation might play a role in the development of endometrial hyperplasia.
Collapse
Affiliation(s)
- Xin Li
- 1. Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden. ; 2. Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China; ; 3. Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Bano Pishdari
- 1. Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peng Cui
- 4. Department of Integrative Medicine and Neurobiology, State Key Lab of Medical Neurobiology, Shanghai Medical College and Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institute of Brain Science, Fudan University, Shanghai, China
| | - Min Hu
- 1. Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hong-Ping Yang
- 2. Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China; ; 3. Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yan-Rong Guo
- 2. Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China; ; 3. Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Hong-Yuan Jiang
- 2. Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China; ; 3. Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yi Feng
- 1. Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden. ; 4. Department of Integrative Medicine and Neurobiology, State Key Lab of Medical Neurobiology, Shanghai Medical College and Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institute of Brain Science, Fudan University, Shanghai, China
| | - Håkan Billig
- 1. Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ruijin Shao
- 1. Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
65
|
Conte F, Banting L, Teede HJ, Stepto NK. Mental health and physical activity in women with polycystic ovary syndrome: a brief review. Sports Med 2015; 45:497-504. [PMID: 25430602 PMCID: PMC4382527 DOI: 10.1007/s40279-014-0291-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This review was designed to consider the available literature concerning mental health and physical activity in women with polycystic ovary syndrome (PCOS). A systematic approach was taken and two electronic databases (PubMed and EBSCO Research articles published between 1970 and 2013) were searched in 2013 to inform a narrative review. Inclusion criteria encompassed requirements for the research to involve a physical activity intervention and assessment of mental health outcomes in women with PCOS. Seven articles considered mental health outcomes and physical activity interventions for women with PCOS. The results demonstrated positive outcomes following physical activity intervention for health-related quality of life, depression, and anxiety. Only one paper reported the independent effects of physical activity on mental health. All other interventions included multi-factor lifestyle interventions or did not establish a control group. Physical activity is likely to be beneficial to the mental health of women with PCOS; however, more research is required to establish the nature of the relationship between physical activity and mental health outcomes.
Collapse
Affiliation(s)
- Francesca Conte
- Institute of Sport, Exercise and Active Living, Victoria University, Footscray Park Campus, PO Box 14428, Melbourne, VIC, 8001, Australia
| | | | | | | |
Collapse
|
66
|
Day FR, Hinds DA, Tung JY, Stolk L, Styrkarsdottir U, Saxena R, Bjonnes A, Broer L, Dunger DB, Halldorsson BV, Lawlor DA, Laval G, Mathieson I, McCardle WL, Louwers Y, Meun C, Ring S, Scott RA, Sulem P, Uitterlinden AG, Wareham NJ, Thorsteinsdottir U, Welt C, Stefansson K, Laven JSE, Ong KK, Perry JRB. Causal mechanisms and balancing selection inferred from genetic associations with polycystic ovary syndrome. Nat Commun 2015; 6:8464. [PMID: 26416764 PMCID: PMC4598835 DOI: 10.1038/ncomms9464] [Citation(s) in RCA: 258] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 08/24/2015] [Indexed: 02/07/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common reproductive disorder in women, yet there is little consensus regarding its aetiology. Here we perform a genome-wide association study of PCOS in up to 5,184 self-reported cases of White European ancestry and 82,759 controls, with follow-up in a further ∼2,000 clinically validated cases and ∼100,000 controls. We identify six signals for PCOS at genome-wide statistical significance (P<5 × 10(-8)), in/near genes ERBB4/HER4, YAP1, THADA, FSHB, RAD50 and KRR1. Variants in/near three of the four epidermal growth factor receptor genes (ERBB2/HER2, ERBB3/HER3 and ERBB4/HER4) are associated with PCOS at or near genome-wide significance. Mendelian randomization analyses indicate causal roles in PCOS aetiology for higher BMI (P=2.5 × 10(-9)), higher insulin resistance (P=6 × 10(-4)) and lower serum sex hormone binding globulin concentrations (P=5 × 10(-4)). Furthermore, genetic susceptibility to later menopause is associated with higher PCOS risk (P=1.6 × 10(-8)) and PCOS-susceptibility alleles are associated with higher serum anti-Müllerian hormone concentrations in girls (P=8.9 × 10(-5)). This large-scale study implicates an aetiological role of the epidermal growth factor receptors, infers causal mechanisms relevant to clinical management and prevention, and suggests balancing selection mechanisms involved in PCOS risk.
Collapse
Affiliation(s)
- Felix R. Day
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | | | | | - Lisette Stolk
- Department of Internal Medicine, Erasmus MC, Rotterdam 3015 GE, The Netherlands
| | | | - Richa Saxena
- Department of Anaesthesia and Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Andrew Bjonnes
- Department of Anaesthesia and Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Linda Broer
- Department of Internal Medicine, Erasmus MC, Rotterdam 3015 GE, The Netherlands
| | - David B. Dunger
- Department of Paediatrics, University of Cambridge School of Clinical Medicine, Box 181, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Bjarni V. Halldorsson
- deCODE Genetics/Amgen, Sturlugata 8, IS-101 Reykjavik, Iceland
- Institute of Biomedical and Neural Engineering, School of Science and Engineering, Reykjavík University, Menntavegur 1, 101 Reykjavík, Iceland
| | - Debbie A. Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol BS8 2BN, UK
- School of Social and Community Medicine, University of Bristol, Oakfield House, Bristol BS8 2BN, UK
| | - Guillaume Laval
- Human Evolutionary Genetics, CNRS URA3012 Institut Pasteur, 28 rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Iain Mathieson
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Wendy L. McCardle
- School of Social and Community Medicine, University of Bristol, Oakfield House, Bristol BS8 2BN, UK
| | - Yvonne Louwers
- Division of Reproductive Medicine, Department of Obstetrics and Gynaecology, Erasmus MC, Rotterdam, 3015 GE, The Netherlands
| | - Cindy Meun
- Division of Reproductive Medicine, Department of Obstetrics and Gynaecology, Erasmus MC, Rotterdam, 3015 GE, The Netherlands
| | - Susan Ring
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol BS8 2BN, UK
- School of Social and Community Medicine, University of Bristol, Oakfield House, Bristol BS8 2BN, UK
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Patrick Sulem
- deCODE Genetics/Amgen, Sturlugata 8, IS-101 Reykjavik, Iceland
| | | | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen, Sturlugata 8, IS-101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, IS-101 Reykjavik, Iceland
| | - Corrine Welt
- Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| | - Kari Stefansson
- deCODE Genetics/Amgen, Sturlugata 8, IS-101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, IS-101 Reykjavik, Iceland
| | - Joop S. E. Laven
- Division of Reproductive Medicine, Department of Obstetrics and Gynaecology, Erasmus MC, Rotterdam, 3015 GE, The Netherlands
| | - Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Department of Paediatrics, University of Cambridge School of Clinical Medicine, Box 181, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - John R. B. Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| |
Collapse
|
67
|
Cui P, Li X, Wang X, Feng Y, Lin JF, Billig H, Shao R. Lack of cyclical fluctuations of endometrial GLUT4 expression in women with polycystic ovary syndrome: Evidence for direct regulation of GLUT4 by steroid hormones. BBA CLINICAL 2015; 4:85-91. [PMID: 26675316 PMCID: PMC4661728 DOI: 10.1016/j.bbacli.2015.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/11/2015] [Accepted: 08/18/2015] [Indexed: 01/06/2023]
Abstract
Background Determination of the role of steroid hormones in expression and regulation of endometrial glucose transport 4 (GLUT4) in humans is important for understanding endometrial disorders such as polycystic ovary syndrome (PCOS), a common hormone-imbalance disease. Methods Endometrial biopsy samples were collected from non-PCOS patients with regular menstrual cycles or with hyperplasia and from PCOS patients with or without hyperplasia. In addition, endometrial tissues from postmenopausal women were incubated with human chorionic gonadotropin (hCG, 10 IU/ml), 17β-estradiol (E2, 10 nM), progesterone (P4, 100 nM), or a combination of E2 and P4 for 24 h. The expression of GLUT4 was measured at the mRNA level using quantitative real-time polymerase chain reaction (qRT-PCR) and at the protein level using Western blot analysis and immunohistochemistry. Results A cyclical change in GLUT4 expression pattern was observed in non-PCOS patients, and a high level of GLUT4 expression was seen in the proliferative phase compared to the secretory phase. Low levels of GLUT4 expression were found in PCOS patients compared to menstrual cycle phase-matched non-PCOS patients, and there was no significant change in GLUT4 expression in PCOS patients during the menstrual cycle. GLUT4 was localized in both epithelial and stromal cells, with notable changes in epithelial cells. We postulate that decreased GLUT4 expression might be regulated by steroid hormones. In support of this, we showed that in cultured endometrial tissues hCG and E2 alone had no effect on GLUT4 expression. However, P4 alone and P4 in combination with E2 decreased GLUT4 expression. Compared with non-PCOS controls, PCOS patients with endometrial hyperplasia exhibited decreased GLUT4 expression in particular in the epithelial cells. Conclusion We conclude that P4 can induce changes in endometrial GLUT4 expression during the menstrual cycle and that abnormal hormonal conditions such as PCOS disrupt normal patterns of GLUT4 expression in endometrial cells.
Collapse
Affiliation(s)
- Peng Cui
- Department of Integrative Medicine and Neurobiology, State Key Lab of Medical Neurobiology, Shanghai Medical College and Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institute of Brain Science, Fudan University, 200032 Shanghai, China
| | - Xin Li
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden ; Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 200011, Shanghai China ; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, 200011 Shanghai, China
| | - Xiaoqin Wang
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden ; BIOMATCELL VINN Excellence Center of Biomaterials and Cell Therapy, Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, Gothenburg 40530, Sweden
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, State Key Lab of Medical Neurobiology, Shanghai Medical College and Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institute of Brain Science, Fudan University, 200032 Shanghai, China ; Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Jin-Fang Lin
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 200011, Shanghai China ; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, 200011 Shanghai, China
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Ruijin Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| |
Collapse
|
68
|
Palomba S, Santagni S, Falbo A, La Sala GB. Complications and challenges associated with polycystic ovary syndrome: current perspectives. Int J Womens Health 2015; 7:745-63. [PMID: 26261426 PMCID: PMC4527566 DOI: 10.2147/ijwh.s70314] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) represents the most common endocrine dysfunction in fertile women and it is considered a heterogeneous and multifaceted disorder, with multiple reproductive and metabolic phenotypes which differently affect the early- and long-term syndrome’s risks. Women with PCOS present an adverse reproductive profile, including a high risk of pregnancy-induced hypertension, preeclampsia, and gestational diabetes mellitus. Patients with PCOS present not only a higher prevalence of classic cardiovascular risk factors, such as hypertension, dyslipidemia, and type-2 diabetes mellitus, but also of nonclassic cardiovascular risk factors, including mood disorders, such as depression and anxiety. Moreover, at the moment, clinical data on cardiovascular morbidity and mortality in women with PCOS are controversial. Finally, women with PCOS show an increased risk of endometrial cancer compared to non-PCOS healthy women, particularly during premenopausal period. Currently, we are unable to clarify if the increased PCOS early- and long-term risks are totally due to PCOS per se or mostly due to obesity, in particular visceral obesity, that characterized the majority of PCOS patients. In any case, the main endocrine and gynecological scientific societies agree to consider women with PCOS at increased risk of obstetric, cardiometabolic, oncology, and psychological complications throughout life, and it is recommended that these women be accurately assessed with periodic follow-up.
Collapse
Affiliation(s)
- Stefano Palomba
- Unit of Obstetrics and Gynecology, Arcispedale Santa Maria Nuova-Scientific Institute of Treatment and Care (IRCCS), Reggio Emilia, Modena, Italy
| | - Susanna Santagni
- Unit of Obstetrics and Gynecology, Arcispedale Santa Maria Nuova-Scientific Institute of Treatment and Care (IRCCS), Reggio Emilia, Modena, Italy
| | - Angela Falbo
- Unit of Obstetrics and Gynecology, Arcispedale Santa Maria Nuova-Scientific Institute of Treatment and Care (IRCCS), Reggio Emilia, Modena, Italy
| | - Giovanni Battista La Sala
- Unit of Obstetrics and Gynecology, Arcispedale Santa Maria Nuova-Scientific Institute of Treatment and Care (IRCCS), Reggio Emilia, Modena, Italy ; Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
69
|
Gharipour M, Sarrafzadegan N, Sadeghi M, Khosravi A, Hoseini M, Khosravi-Boroujeni H, Khaledifar A. The metabolic syndrome and associated lifestyle factors among the Iranian population. Adv Biomed Res 2015; 4:84. [PMID: 26015910 PMCID: PMC4434446 DOI: 10.4103/2277-9175.156645] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 08/07/2013] [Indexed: 11/17/2022] Open
Abstract
Background: This study aims to evaluate the impact of the Isfahan Healthy Heart Program (IHHP) interventions concerning healthy behavior, on the prevalence of the metabolic syndrome (MetS) and its components in the Iranian population. Materials and Methods: The IHHP targeted the population at large in three districts in central Iran from 2000 to 2007. Numerous interventional activities were performed to improve lifestyle. The main intervention strategies were public education through mass media, intersectional cooperation, health professional education, marketing and organizational development, legislation and policy development, as well as research and evaluation. MetS was defined based on the Adult Treatment Panel (ATP) III definition. The logistic regression method was applied to explore the relationship between lifestyle factors with components of metabolic risk factors. Results: The mean age of the participants was 44.68 ± 14.43 years in 2001. The mean values of the MetS components differed from 2001 to 2007. The mean of systolic blood pressure (SBP) decreased from 126.7 ± 22.31 to 124.21 ± 20.0 and from 129.47 ± 23.08 to 126.26 ± 21.88 among females in both the intervention and reference areas. Similar changes were observed among males. The mean diastolic blood pressure (DBP) and triglycerides decreased significantly in the intervention area and increased significantly in the reference area in both sexes. High density protein cholesterol (HDL-C) was decreased in both sexes, from 2001 to 2007, in both areas. A strong relationship between tobacco control with high SBP and hypertriglyceridemia was found (P < 0.01). Conclusion: Lifestyle improvement programs could be useful to improve the MetS status among men and women.
Collapse
Affiliation(s)
- Mojgan Gharipour
- Department of Metabolic Syndrome, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoumeh Sadeghi
- Cardiac Rehabilitation Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Khosravi
- Department of Research, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Hoseini
- Intervenetional Cardiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khosravi-Boroujeni
- Hypertension Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arsalan Khaledifar
- Department of Cardiology, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
70
|
Dennett CC, Simon J. The role of polycystic ovary syndrome in reproductive and metabolic health: overview and approaches for treatment. Diabetes Spectr 2015; 28:116-20. [PMID: 25987810 PMCID: PMC4433074 DOI: 10.2337/diaspect.28.2.116] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
| | - Judy Simon
- University of Washington Department of Medicine Roosevelt Clinic, Seattle, WA
- Mind Body Nutrition, Bellevue, WA
| |
Collapse
|
71
|
Orio F, Muscogiuri G, Palomba S. Could the Mediterranean diet be effective in women with polycystic ovary syndrome? A proof of concept. Eur J Clin Nutr 2015; 69:974. [PMID: 25828622 DOI: 10.1038/ejcn.2015.53] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- F Orio
- Department of Sports Science and Wellness, "Parthenope" University Naples, and Endocrinology and Diabetology, Fertility Techniques Structure, University Hospital "S. Giovanni di Dio e Ruggi d'Aragona", Largo Città d'Ippocrate, Salerno, Italy
| | - G Muscogiuri
- Department of Clinical Medicine and Surgery, Section of Endocrinology, University "Federico II", Naples and Rionero in Vulture, Potenza, Italy
| | - S Palomba
- Department of Obstetrics and Gynecology, Arcispedale S. Maria Nuova of Reggio Emilia, IRCCS, Reggio Emilia, Italy
| |
Collapse
|
72
|
Identify underlying cause of hirsutism and individualize treatment as required. DRUGS & THERAPY PERSPECTIVES 2014. [DOI: 10.1007/s40267-014-0159-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
73
|
Shao R, Li X, Billig H. Promising clinical practices of metformin in women with PCOS and early-stage endometrial cancer. BBA CLINICAL 2014; 2:7-9. [PMID: 26674650 PMCID: PMC4633941 DOI: 10.1016/j.bbacli.2014.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 06/24/2014] [Accepted: 07/12/2014] [Indexed: 12/01/2022]
Abstract
Young women with polycystic ovary syndrome (PCOS) have a high risk of developing endometrial cancer (EC). There is an urgent need for non-surgical prevention and treatment strategies for these patients who fail to respond to progesterone treatment and wish to preserve their fertility. Recently, we have reported that the combined treatment with metformin and progesterone-based oral contraceptives has successfully reversed the early-stage EC into normal endometria in addition to improvement of insulin resistance in women with PCOS. More importantly, one of these treated women has successfully delivered a healthy newborn baby. However, before such treatment can be recommended to the clinical practice, the molecular basis of metformin in the endometrium under physiological and pathological conditions must be elucidated.
Collapse
Affiliation(s)
- Ruijin Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Xin Li
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden ; Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| |
Collapse
|
74
|
Abstract
BACKGROUND Hirsutism has a relatively high prevalence among women. Depending upon societal and ethnic norms, it can cause significant psychosocial distress. Importantly, hirsutism may be associated with underlying disorders and co-morbidities. Hirsutism should not simply be looked upon as an issue of cosmesis. Patients require appropriate evaluation so that underlying etiologies and associated sequelae are recognized and managed. Treatment of hirsutism often requires a multidisciplinary approach, and a variety of physical or pharmacologic modalities can be employed. Efficacy of these therapies is varied and depends, among other things, upon patient factors including the underlying etiology, hormonal drive, and local tissue sensitivity to androgens. OBJECTIVE The objective of this paper is to review and summarize current evidence evaluating the efficacy of various treatment modalities for hirsutism in premenopausal women. METHODS Online databases were searched to identify all relevant prior systematic reviews and meta-analyses as well as recently published (2012-present) randomized controlled trials (RCTs) on hirsutism treatment. RESULTS Four recently published RCTs met criteria for inclusion in our review. In addition, one meta-analysis and one systematic review/treatment guideline were identified in the recent literature. Physical modalities and oral contraceptive pills (OCPs) remain first-line treatments. Evidence supports the use of electrolysis for permanent hair removal in localized areas and lasers (particularly alexandrite and diode lasers) for permanent hair reduction. Topical eflornithine can be used as monotherapy for mild hirsutism and as an adjunct therapy with lasers or pharmacotherapy in more severe cases. Combined OCPs as a class are superior to placebo; however, antiandrogenic and low-dose neutral OCPs may be slightly more efficacious in improving hirsutism compared with other types of OCPs. Antiandrogens are indicated for moderate to severe hirsutism, with spironolactone being the first-line antiandrogen and finasteride and cyproterone acetate being second-line antiandrogens. Due to its risk for hepatotoxicity, flutamide is not considered a first-line therapy. If used, the lowest effective dose should be administered with careful monitoring of liver enzymes. Monotherapy with an insulin sensitizer does not significantly improve hirsutism. While insulin sensitizers improve important metabolic and endocrine aberrations in polycystic ovary syndrome, they are not recommended when hirsutism is the sole indication for use. Lifestyle modification counseling is recommended. Gonadotropin-releasing hormone analogs and glucocorticoids are only recommended in specific circumstances. Additional therapies without sufficient supportive evidence of efficacy are ovarian surgery, statins (HMG-CoA reductase inhibitors), and vitamin D supplementation. LIMITATIONS In general, most therapies garner recommendations that are weak (where the estimates of benefits versus risks of therapy are either closely balanced or uncertain) and are based on low- to moderate-quality evidence. CONCLUSIONS Risks and benefits of treatment must be carefully considered and discussed with the patient. Expectations for efficacy should be appropriately set. A minimum of 6 months is required to see benefit from pharmacotherapy and lifelong treatment is often necessary for sustained benefit.
Collapse
|
75
|
Progressive Resistance Training in Polycystic Ovary Syndrome: Can Pumping Iron Improve Clinical Outcomes? Sports Med 2014; 44:1197-207. [DOI: 10.1007/s40279-014-0206-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
76
|
Seli E, Babayev E, Collins SC, Nemeth G, Horvath TL. Minireview: Metabolism of female reproduction: regulatory mechanisms and clinical implications. Mol Endocrinol 2014; 28:790-804. [PMID: 24678733 DOI: 10.1210/me.2013-1413] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Female fertility is highly dependent on successful regulation of energy metabolism. Central processes in the hypothalamus monitor the metabolic state of the organism and, together with metabolic hormones, drive the peripheral availability of energy for cellular functions. In the ovary, the oocyte and neighboring somatic cells of the follicle work in unison to achieve successful metabolism of carbohydrates, amino acids, and lipids. Metabolic disturbances such as anorexia nervosa, obesity, and diabetes mellitus have clinically important consequences on human reproduction. In this article, we review the metabolic determinants of female reproduction and their role in infertility.
Collapse
Affiliation(s)
- Emre Seli
- Department of Obstetrics, Gynecology, and Reproductive Sciences (E.S., E.B., S.C., T.L.H.), Yale School of Medicine, New Haven, Connecticut 06520; Department of Obstetrics and Gynecology (G.N., T.L.H.), University of Szeged, Faculty of Medicine, Szeged, Hungary 6701; Department of Comparative Medicine (T.L.H.), Yale School of Medicine, New Haven, Connecticut 06520; and the Department of Neurobiology (T.L.H.), Yale School of Medicine, New Haven, Connecticut 06520
| | | | | | | | | |
Collapse
|
77
|
Louwers YV, Rayner NW, Herrera BM, Stolk L, Groves CJ, Barber TM, Uitterlinden AG, Franks S, Laven JSE, McCarthy MI. BMI-associated alleles do not constitute risk alleles for polycystic ovary syndrome independently of BMI: a case-control study. PLoS One 2014; 9:e87335. [PMID: 24498077 PMCID: PMC3909077 DOI: 10.1371/journal.pone.0087335] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 12/20/2013] [Indexed: 12/15/2022] Open
Abstract
Introduction Polycystic Ovary Syndrome (PCOS) has a strong genetic background and the majority of patients with PCOS have elevated BMI levels. The aim of this study was to determine to which extent BMI-increasing alleles contribute to risk of PCOS when contemporaneous BMI is taken into consideration. Methods Patients with PCOS and controls were recruited from the United Kingdom (563 cases and 791 controls) and The Netherlands (510 cases and 2720 controls). Cases and controls were of similar BMI. SNPs mapping to 12 BMI-associated loci which have been extensively replicated across different ethnicities, i.e., BDNF, FAIM2, ETV5, FTO, GNPDA2, KCTD15, MC4R, MTCH2, NEGR1, SEC16B, SH2B1, and TMEM18, were studied in association with PCOS within each cohort using the additive genetic model followed by a combined analysis. A genetic allelic count risk score model was used to determine the risk of PCOS for individuals carrying increasing numbers of BMI-increasing alleles. Results None of the genetic variants, including FTO and MC4R, was associated with PCOS independently of BMI in the meta-analysis. Moreover, no differences were observed between cases and controls in the number of BMI-risk alleles present and no overall trend across the risk score groups was observed. Conclusion In this combined analysis of over 4,000 BMI-matched individuals from the United Kingdom and the Netherlands, we observed no association of BMI risk alleles with PCOS independent of BMI.
Collapse
Affiliation(s)
- Yvonne V. Louwers
- Department of Obstetrics and Gynecology, Subdivision of Reproductive Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Nigel W. Rayner
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Blanca M. Herrera
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Lisette Stolk
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Christopher J. Groves
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Thomas M. Barber
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Andre G. Uitterlinden
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Joop S. E. Laven
- Department of Obstetrics and Gynecology, Subdivision of Reproductive Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- * E-mail: (MIM); (JSEL)
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- * E-mail: (MIM); (JSEL)
| |
Collapse
|
78
|
Li X, Guo YR, Lin JF, Feng Y, Billig H, Shao R. Combination of Diane-35 and Metformin to Treat Early Endometrial Carcinoma in PCOS Women with Insulin Resistance. J Cancer 2014; 5:173-81. [PMID: 24563672 PMCID: PMC3931265 DOI: 10.7150/jca.8009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 01/14/2014] [Indexed: 01/02/2023] Open
Abstract
Background: Young women with polycystic ovary syndrome (PCOS) have a high risk of developing endometrial carcinoma. There is a need for the development of new medical therapies that can reduce the need for surgical intervention so as to preserve the fertility of these patients. The aim of the study was to describe and discuss cases of PCOS and insulin resistance (IR) women with early endometrial carcinoma while being co-treated with Diane-35 and metformin. Methods: Five PCOS-IR women who were scheduled for diagnosis and therapy for early endometrial carcinoma were recruited. The hospital records and endometrial pathology reports were reviewed. All patients were co-treated with Diane-35 and metformin for 6 months to reverse the endometrial carcinoma and preserve their fertility. Before, during, and after treatment, endometrial biopsies and blood samples were obtained and oral glucose tolerance tests were performed. Endometrial pathology was evaluated. Body weight (BW), body mass index (BMI), follicle-stimulating hormone (FSH), luteinizing hormone (LH), total testosterone (TT), sex hormone-binding globulin (SHBG), free androgen index (FAI), insulin area under curve (IAUC), and homeostasis model assessment of insulin resistance (HOMA-IR) were determined. Results: Clinical stage 1a, low grade endometrial carcinoma was confirmed before treatment. After 6 months of co-treatment, all patients showed normal epithelia. No evidence of atypical hyperplasia or endometrial carcinoma was found. Co-treatment resulted in significant decreases in BW, BMI, TT, FAI, IAUC, and HOMA-IR in parallel with a significant increase in SHBG. There were no differences in the FSH and LH levels after co-treatment. Conclusions: Combined treatment with Diane-35 and metformin has the potential to revert the endometrial carcinoma into normal endometrial cells in PCOS-IR women. The cellular and molecular mechanisms behind this effect merit further investigation.
Collapse
Affiliation(s)
- Xin Li
- 1. Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China ; 2. Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Yan-Rong Guo
- 1. Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Jin-Fang Lin
- 1. Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Yi Feng
- 2. Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden ; 3. Department of Integrative Medicine and Neurobiology, State Key Lab of Medical Neurobiology, Shanghai Medical College and Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institute of Brain Science, Fudan University, Shanghai 200032, China
| | - Håkan Billig
- 2. Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Ruijin Shao
- 2. Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| |
Collapse
|
79
|
Li X, Feng Y, Lin JF, Billig H, Shao R. Endometrial progesterone resistance and PCOS. J Biomed Sci 2014; 21:2. [PMID: 24405633 PMCID: PMC3917599 DOI: 10.1186/1423-0127-21-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/06/2014] [Indexed: 01/25/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a state of altered steroid hormone production and activity. Chronic estrogen exposure or lack of progesterone due to ovarian dysfunction can result in endometrial hyperplasia and carcinoma. A key contributor to our understanding of progesterone as a critical regulator for normal uterine function has been the elucidation of progesterone receptor (PR) expression, regulation, and signaling pathways. Several human studies indicate that PR-mediated signaling pathways in the nucleus are associated with progesterone resistance in women with PCOS. The aim of this review is to provide an overview of endometrial progesterone resistance in women with PCOS; to present the PR structure, its different isoforms, and their expression in the endometrium; to illustrate the possible regulation of PR and PR-mediated signaling in progesterone resistance in women with PCOS; and to discuss current clinical treatments for atypical endometrial hyperplasia and endometrial carcinoma in women with PCOS and accompanying progesterone resistance.
Collapse
Affiliation(s)
| | | | | | | | - Ruijin Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden.
| |
Collapse
|
80
|
Haqq L, McFarlane J, Dieberg G, Smart N. Effect of lifestyle intervention on the reproductive endocrine profile in women with polycystic ovarian syndrome: a systematic review and meta-analysis. Endocr Connect 2014; 3:36-46. [PMID: 24488490 PMCID: PMC3938041 DOI: 10.1530/ec-14-0010] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Polycystic ovarian syndrome (PCOS) affects 18-22% of women at reproductive age. We conducted a systematic review and meta-analysis evaluating the expected benefits of lifestyle (exercise plus diet) interventions on the reproductive endocrine profile in women with PCOS. Potential studies were identified by systematically searching PubMed, CINAHL and the Cochrane Controlled Trials Registry (1966-April 30, 2013) systematically using key concepts of PCOS. Significant improvements were seen in women receiving lifestyle intervention vs usual care in follicle-stimulating hormone (FSH) levels, mean difference (MD) 0.39 IU/l (95% CI 0.09 to 0.70, P=0.01), sex hormone-binding globulin (SHBG) levels, MD 2.37 nmol/l (95% CI 1.27 to 3.47, P<0.0001), total testosterone levels, MD -0.13 nmol/l (95% CI -0.22 to -0.03, P=0.008), androstenedione levels, MD -0.09 ng/dl (95% CI -0.15 to -0.03, P=0.005), free androgen index (FAI) levels, MD -1.64 (95% CI -2.94 to -0.35, P=0.01) and Ferriman-Gallwey (FG) score, MD -1.01 (95% CI -1.54 to -0.48, P=0.0002). Significant improvements were also observed in women who received exercise-alone intervention vs usual care in FSH levels, MD 0.42 IU/l (95% CI 0.11 to 0.73, P=0.009), SHBG levels, MD 3.42 nmol/l (95% CI 0.11 to 6.73, P=0.04), total testosterone levels, MD -0.16 nmol/l (95% CI -0.29 to -0.04, P=0.01), androstenedione levels, MD -0.09 ng/dl (95% CI -0.16 to -0.03, P=0.004) and FG score, MD -1.13 (95% CI -1.88 to -0.38, P=0.003). Our analyses suggest that lifestyle (diet and exercise) intervention improves levels of FSH, SHBG, total testosterone, androstenedione and FAI, and FG score in women with PCOS.
Collapse
Affiliation(s)
| | | | | | - Neil Smart
- correspondence should be addressed to N Smart
| |
Collapse
|
81
|
Panidis D, Tziomalos K, Papadakis E, Vosnakis C, Chatzis P, Katsikis I. Lifestyle intervention and anti-obesity therapies in the polycystic ovary syndrome: impact on metabolism and fertility. Endocrine 2013; 44:583-590. [PMID: 23625194 DOI: 10.1007/s12020-013-9971-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 04/23/2013] [Indexed: 12/17/2022]
Abstract
Obesity is frequently present in patients with polycystic ovary syndrome (PCOS) and plays an important role in the pathogenesis of the metabolic, endocrine, and reproductive abnormalities associated with this syndrome. We aimed to summarize the effects of lifestyle changes and anti-obesity pharmacotherapy in patients with PCOS. We reviewed the literature regarding the effects of lifestyle changes and anti-obesity agents on the metabolic and endocrine abnormalities of PCOS. Lifestyle changes, including diet, exercise, and behavioral modification, appear to improve the metabolic and reproductive abnormalities of overweight and obese patients with PCOS. Therefore, lifestyle changes appear to represent the first-line management for all overweight and obese patients with PCOS. However, the optimal composition of diet and the optimal type of exercise in these patients are unknown. Anti-obesity agents that have been studied in PCOS include orlistat, sibutramine, and rimonabant. However, the latter two agents have been withdrawn from the market because of side effects. Long-term studies with orlistat in overweight and obese diabetic patients showed greater weight loss and metabolic and cardiovascular benefits than those achieved with lifestyle changes alone. However, there are limited data on the efficacy of orlistat in women with PCOS. In conclusion, lifestyle changes (diet, exercise and behavioral modification), particularly when combined with anti-obesity agents, exert beneficial effects on the endocrine abnormalities of obese patients with PCOS and improve metabolic parameters.
Collapse
Affiliation(s)
- Dimitrios Panidis
- Division of Endocrinology and Human Reproduction, Second Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | | | | | | | | | | |
Collapse
|
82
|
Johansson J, Stener-Victorin E. Polycystic ovary syndrome: effect and mechanisms of acupuncture for ovulation induction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:762615. [PMID: 24073009 PMCID: PMC3773899 DOI: 10.1155/2013/762615] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/23/2013] [Indexed: 12/22/2022]
Abstract
Polycystic ovary syndrome (PCOS), the most common endocrine disorder among women of reproductive age, is characterized by the coexistence of hyperandrogenism, ovulatory dysfunction, and polycystic ovaries (PCO). PCOS also represents the largest part of female oligoovulatory infertility, and the management of ovulatory and menstrual dysfunction, comprises a third of the high costs of PCOS treatment. Current pharmacological and surgical treatments for reproductive symptoms are effective, however, associated with negative side effects, such as cardiovascular complications and multiple pregnancies. For menstrual irregularities and ovulation induction in women with PCOS, acupuncture has indicated beneficial effects. This review will focus on the results from randomized controlled acupuncture trials for regulation of menstrual dysfunction and for inducing ovulation in women with PCOS although there are uncontrolled trials with nonetheless interesting results. Animal experimental studies will be further discussed when they can provide a more mechanistic explanatory view.
Collapse
Affiliation(s)
- Julia Johansson
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Box 434, 405 30 Gothenburg, Sweden
| | - Elisabet Stener-Victorin
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Box 434, 405 30 Gothenburg, Sweden
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| |
Collapse
|
83
|
Li Y, Ma H, Zhang Y, Kuang H, Ng EHY, Hou L, Wu X. Effect of berberine on insulin resistance in women with polycystic ovary syndrome: study protocol for a randomized multicenter controlled trial. Trials 2013; 14:226. [PMID: 23866924 PMCID: PMC3722087 DOI: 10.1186/1745-6215-14-226] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 06/28/2013] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Insulin resistance and hyperinsulinemia play a key role in the pathogenesis of polycystic ovary syndrome (PCOS), which is characterized by hyperandrogenism, ovulatory dysfunction, and presence of polycystic ovaries on pelvic scanning. Insulin resistance is significantly associated with the long-term risks of metabolic syndrome and cardiovascular disease. Berberine has effects on insulin resistance but its use in women with PCOS has not been fully investigated. In this paper, we present a research design evaluating the effects of berberine on insulin resistance in women with PCOS. METHODS/DESIGN This is a multicenter, randomized, placebo-controlled and double-blind trial. A total of 120 patients will be enrolled in this study and will be randomized into two groups. Berberine or placebo will be taken orally for 12 weeks. The primary outcome is the whole body insulin action assessed with the hyperinsulinemic-euglycemic clamp. DISCUSSION We postulate that women with PCOS will have improved insulin resistance following berberine administration. TRIAL REGISTRATION This study is registered at ClinicalTrials.gov, NCT01138930.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | | | | | | | | | | | | |
Collapse
|
84
|
Kim C, Pi-Sunyer X, Barrett-Connor E, Stentz FB, Murphy MB, Kong S, Nan B, Kitabchi AE. Sex hormone binding globulin and sex steroids among premenopausal women in the diabetes prevention program. J Clin Endocrinol Metab 2013; 98:3049-57. [PMID: 23709655 PMCID: PMC3701280 DOI: 10.1210/jc.2013-1341] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
CONTEXT It is unknown whether intensive lifestyle modification (ILS) or metformin changes sex steroids among premenopausal women without a history of polycystic ovarian syndrome (PCOS). OBJECTIVES We examined 1-year intervention impact on sex steroids (estradiol, testosterone, dehydroepiandrosterone, and androstenedione [A4]) and SHBG and differences by race/ethnicity. PARTICIPANTS A subgroup of Diabetes Prevention Program participants who were premenopausal, not using estrogen, without a history of PCOS or irregular menses, and who reported non-Hispanic white (NHW), Hispanic, or African-American race/ethnicity (n = 301). INTERVENTIONS Randomization arms were 1) ILS with the goals of weight reduction of 7% of initial weight and 150 minutes per week of moderate intensity exercise, 2) metformin 850 mg twice a day, or 3) placebo. RESULTS Neither intervention changed sex steroids compared to placebo. ILS, but not metformin, increased median SHBG by 3.1 nmol/L (~11%) compared to decreases of 1.1 nmol/L in the placebo arm (P < .05). This comparison remained significant after adjustment for changes in covariates including waist circumference. However, associations with glucose were not significant. Median baseline A4 was lower in Hispanics compared to NHWs (5.7 nmol/L vs 6.5 nmol/L, P < .05) and increases in A4 were greater in Hispanics compared to NHWs (3.0 nmol/ vs 1.2 nmol/L, P < .05), and these differences did not differ significantly by intervention arm. No other racial/ethnic differences were significant. CONCLUSIONS Among premenopausal glucose-intolerant women, no intervention changed sex steroids. ILS increased SHBG, although associations with glucose were not significant. SHBG and sex steroids were similar by race/ethnicity, with the possible exception of lower baseline A4 levels in Hispanics compared to NHWs.
Collapse
Affiliation(s)
- Catherine Kim
- University of Michigan, Ann Arbor, Michigan 48109-2800, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Malik SM, Traub ML. Defining the role of bariatric surgery in polycystic ovarian syndrome patients. World J Diabetes 2012; 3:71-9. [PMID: 22532886 PMCID: PMC3334389 DOI: 10.4239/wjd.v3.i4.71] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/21/2012] [Accepted: 04/10/2012] [Indexed: 02/05/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrine disorder in women. To meet PCOS criteria, women must have a combination of hyperandrogenism, anovulation and ultrasound findings. Almost 10% of all reproductive age women worldwide show signs of PCOS. Although women often seek care for gynecological or body image concerns, many PCOS women are at risk for metabolic syndrome (MS). Many of the metabolic consequences are overlooked and undertreated by physicians because these patients tend to be young, reproductive age women. MS and obesity coexist commonly with PCOS. These young women are predisposed to glucose abnormalities and ultimately diabetes mellitus, dyslipidemia and eventually cardiovascular disease. Bariatric surgery can be an effective means of weight loss in PCOS women. Surgical techniques have become safer and less invasive over time and have been found to be effective in achieving significant weight loss. Surgical options have also increased, giving patients more choices. Bariatric surgery may prevent or reverse metabolic syndrome. Bariatric surgery may also have reproductive benefits in PCOS patients. Although bariatric surgery has historically been performed in older, reproductive aged women, it has recently gained favor in adolescents as well. This is of particular importance due to the prevalence of both PCOS and MS in adolescents. Treatment of PCOS and MS certainly requires a combination of medical therapy, psychological support and lifestyle modifications. These treatments are difficult and often frustrating for patients and physicians. Bariatric surgery can be effective in achieving significant weight loss, restoration of the hypothalamic pituitary axis, reduction of cardiovascular risk and even in improving pregnancy outcomes. Ultimately, bariatric surgery should be considered part of the treatment in PCOS women, especially in those with MS.
Collapse
Affiliation(s)
- Shaveta M Malik
- Shaveta M Malik, Michael L Traub, Department of Obstetrics and Gynecology, Staten Island University Hospital, Staten Island, NY 10305, United States
| | | |
Collapse
|
86
|
Kaidar-Person O, Bar-Sela G, Person B. The two major epidemics of the twenty-first century: obesity and cancer. Obes Surg 2012; 21:1792-7. [PMID: 21842287 DOI: 10.1007/s11695-011-0490-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It is a well-known fact that severe obesity is associated with the metabolic syndrome, type 2 diabetes, cardiovascular disease, hypertension, and other diseases. Epidemiological studies have suggested that obesity is also associated with increased risk of several cancer types. The number of people who are suffering from severe obesity is growing, and clinical data suggest that severely obese patients belong to a unique population with regards to risk, efficacy of screening, and cancer treatment. This review will point out the potential mechanism linking obesity and cancer and will discuss several challenges in various treatment modalities of cancer in obese patients.
Collapse
Affiliation(s)
- Orit Kaidar-Person
- Division of Oncology, Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | |
Collapse
|
87
|
Abu Hashim H. Clomiphene citrate alternatives for the initial management of polycystic ovary syndrome: an evidence-based approach. Arch Gynecol Obstet 2012; 285:1737-45. [DOI: 10.1007/s00404-012-2261-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 02/16/2012] [Indexed: 11/29/2022]
|
88
|
Roos N, Kieler H, Sahlin L, Ekman-Ordeberg G, Falconer H, Stephansson O. Risk of adverse pregnancy outcomes in women with polycystic ovary syndrome: population based cohort study. BMJ 2011; 343:d6309. [PMID: 21998337 PMCID: PMC3192872 DOI: 10.1136/bmj.d6309] [Citation(s) in RCA: 193] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To study the risk of adverse pregnancy outcomes in women with polycystic ovary syndrome, taking into account maternal characteristics and assisted reproductive technology. DESIGN Population based cohort study. SETTING Singleton births registered in the Swedish medical birth register between 1995 and 2007. PARTICIPANTS By linkage with the Swedish patient register, 3787 births among women with a diagnosis of polycystic ovary syndrome and 1,191,336 births among women without such a diagnosis. MAIN OUTCOME MEASURES Risk of adverse pregnancy outcomes (gestational diabetes, pre-eclampsia, preterm birth, stillbirth, neonatal death, low Apgar score (<7 at five minutes), meconium aspiration, large for gestational age, macrosomia, small for gestational age), adjusted for maternal characteristics (body mass index, age), socioeconomic factors (educational level, and cohabitating with infant's father), and assisted reproductive technology. RESULTS Women with polycystic ovary syndrome were more often obese and more commonly used assisted reproductive technology than women without such a diagnosis (60.6% v 34.8% and 13.7% v 1.5%). Polycystic ovary syndrome was strongly associated with pre-eclampsia (adjusted odds ratio 1.45, 95% confidence interval 1.24 to 1.69) and very preterm birth (2.21, 1.69 to 2.90) and the risk of gestational diabetes was more than doubled (2.32, 1.88 to 2.88). Infants born to mothers with polycystic ovary syndrome were more prone to be large for gestational age (1.39, 1.19 to 1.62) and were at increased risk of meconium aspiration (2.02, 1.13 to 3.61) and having a low Apgar score (<7) at five minutes (1.41, 1.09 to 1.83). CONCLUSIONS Women with polycystic ovary syndrome are at increased risk of adverse pregnancy and birth outcomes that cannot be explained by assisted reproductive technology. These women may need increased surveillance during pregnancy and parturition.
Collapse
Affiliation(s)
- Nathalie Roos
- Department of Women's and Children's Health, Division of Obstetrics and Gynaecology, Karolinska Institutet, Karolinska University Hospital, H2:01, SE-171 76 Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
89
|
Wilson-Pérez HE, Seeley RJ. The effect of vertical sleeve gastrectomy on a rat model of polycystic ovarian syndrome. Endocrinology 2011; 152:3700-5. [PMID: 21810946 PMCID: PMC3176643 DOI: 10.1210/en.2011-1241] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 07/13/2011] [Indexed: 02/03/2023]
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrine disorder of women of reproductive age. Although some of the primary symptoms of PCOS are reproductive abnormalities, including hyperandrogenism, menstrual dysfunction, and hirsutism, other metabolic disturbances are also common, including obesity and insulin resistance. Women with PCOS who have undergone weight-loss bariatric surgery have reported surprising postoperative benefits beyond weight loss, including resolution of menstrual dysfunction and improvement of hirsutism. Here, we use a chronic dihydrotestosterone (DHT) exposure model of PCOS in female rats and investigate the efficacy of a specific type of bariatric surgery, namely vertical sleeve gastrectomy (VSG), to resolve the reproductive and metabolic disturbances induced by DHT treatment. We find that VSG causes loss of body weight and body fat in DHT-treated rats but does not improve glucose tolerance or restore estrous cyclicity. Although human PCOS patients have shown decreased androgen levels after bariatric surgery, the chronic nature of DHT administration in this rat model both before and after VSG renders this effect impossible in this case. Therefore, the lack of improvement in glucose tolerance and estrous cyclicity may implicate a direct effect of androgen knockdown as a mechanism for the improvements seen in human PCOS patients after bariatric surgery. In addition, the dissociation of body weight loss without improved glucose tolerance suggests that glucose intolerance may be a body weight-independent phenomenon in women with PCOS.
Collapse
|