1
|
Wu S, Wu Y, Fang L, Lu X. Association of insulin resistance surrogates with live birth outcomes in women with polycystic ovary syndrome undergoing in vitro fertilization. BMC Pregnancy Childbirth 2025; 25:25. [PMID: 39799297 PMCID: PMC11724488 DOI: 10.1186/s12884-024-07131-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/31/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Insulin resistance (IR) is a common pathophysiologic feature in patients with polycystic ovary syndrome (PCOS). However, there have been no studies investigating the association of IR surrogates with pregnancy outcomes in women with PCOS undergoing in vitro fertilization (IVF). Therefore, we explored the association between these factors among PCOS patients. METHODS We conducted a retrospective study that included patients with PCOS who underwent IVF at a university-affiliated hospital. Blood samples and physical examinations are collected at reproductive center on fasting in the morning of the 2nd to 4th day of the menstrual cycle prior to medication. We categorized participants into "Non-IR group" (HOMA-IR < 2.2) and "IR group" (HOMA-IR ≥ 2.2). The association of IR surrogates [triglyceride-glucose-body mass index (TyG-BMI), triglyceride-glucose (TyG) and homeostasis model assessment (HOMA-IR)] with IVF outcomes was evaluated by regression model analysis. Moreover, we also performed sensitivity analyses with stratification and interaction tests. The primary outcome variable was the live birth rate. RESULTS A total of 543 PCOS patients were finally included in the study. In all three regression models for the fresh embryo transfer (ET) cycles, all three IR surrogates showed stable negative correlations with live birth rate (in Model III: TyG-BMI OR = 0.99, 95% CI: 0.98 ~ 0.99; TyG OR = 0.47, 95% CI: 0.27 ~ 0.82; HOMA-IR OR = 0.84, 95% CI: 0.72 ~ 0.97; all P < 0.05), and this association was stable across all subgroups of the population (all P-interaction > 0.05). However, this relationship did not exist in frozen-thawed embryo transfer (FET) cycles. Furthermore, our study found that TyG-BMI was superior to TyG and HOMA-IR in predicting the rate of live birth in fresh ET cycles [TyG-BMI: 0.64 (95% CI: 0.58, 0.69) vs. TyG: 0.61 (95% CI: 0.55, 0.67) vs. HOMA-IR: 0.60 (95% CI: 0.55, 0.67)]. CONCLUSIONS Our study revealed that the three IR surrogates (TyG-BMI, TyG and HOMA-IR) were negatively associated with the live birth rates in fresh ET cycles. However, this relationship did not exist in FET cycles. Furthermore, our study found that TyG-BMI was superior to TyG and HOMA-IR in predicting the rate of live birth in fresh ET cycles.
Collapse
Affiliation(s)
- Shenghao Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yanhong Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Lizi Fang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiaosheng Lu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
2
|
Liu H, Shangguan F, Liu F, Guo Y, Yu H, Li H, Su Y, Li Z. Evaluating the effects of time-restricted eating on overweight and obese women with polycystic ovary syndrome: A randomized controlled trial study protocol. PLoS One 2025; 20:e0316333. [PMID: 39787136 PMCID: PMC11717230 DOI: 10.1371/journal.pone.0316333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Time-restricted eating (TRE) manages weight effectively, but choosing how long and what time window remain debatable. Although an 8:00 a.m. to 16:00 p.m. time frame is reported to show positive results in most weight loss trial, its safety and efficacy in overweight and obese women with polycystic ovary syndrome (PCOS) is uncertain. This randomized controlled trial is conducted to evaluate the safety and efficacy of TRE in specific populations. OBJECTIVE This study aims to assess the 6-month effects of TRE on weight change, metabolic improvement, reproductive recovery, and health-related quality of life in overweight and obese women with polycystic ovary syndrome (PCOS), compared to those who did not receive TRE. METHODS This randomized controlled trial will enroll 96 overweight and obese women with polycystic ovary syndrome (PCOS), who will be randomly assigned to either a TRE group (with an eating window from 8:00 a.m. to 16:00 p.m.) or a control group (without eating time restrictions), with 49 participants in each group. Evaluators and data analysts will remain blinded to group allocation throughout the study. The primary outcomes, including changes in weight and body mass index (BMI), will be assessed weekly. Secondary outcomes, encompassing alterations in sex hormones, metabolic parameters, body composition, sleep quality, quality of life, anxiety, and depression, will be evaluated monthly. Compliance and safety will be continuously monitored throughout the study. Additionally, a 6-month follow-up will be conducted at the end of the trial to assess the long-term effects of TRE. Statistical analysis will include the Anderson-Darling test for normality, T-test/Wilcoxon test based on distribution, mixed-effects models for assessing time/group effects, Cox model for time-to-event analysis, repeated ANOVA for change analysis, and sensitivity analysis. All tests will be conducted using appropriate software, with a significance level set at P<0.05. Missing data will be imputed. DISCUSSION The purpose of this study protocol is to further evaluate the effects of TRE in overweight and obese women with PCOS through a randomized controlled trial (RCT). Findings from this study are expected to provide new dietary intervention strategies for overweight and obese PCOS participants. ETHICS AND DISSEMINATION This study has received ethics approval from the Medical Ethics Committee of the University of South China (Number: NHHL027). Participants are included after signing informed consent. Results will be submitted for publication in peer-reviewed journals. TRAIL REGISTRATION Trail registration number: ChiCTR2400086815.
Collapse
Affiliation(s)
- Hui Liu
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Fuliang Shangguan
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Fen Liu
- The First Affliated Hospital, University of South China, Hengyang, Hunan, China
| | - Yu Guo
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Huixi Yu
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hanbing Li
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yinhua Su
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhongyu Li
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
3
|
Perng W, Fitz VW, Salmon K, Hivert MF, Kazemi M, Rifas-Shiman SL, Shifren J, Oken E, Chavarro JE. Prevalence and correlates of diagnosed and probable polycystic ovary syndrome (PCOS) in a cohort of parous women. Am J Epidemiol 2025; 194:114-121. [PMID: 38960722 DOI: 10.1093/aje/kwae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 05/08/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
Correlates of diagnosed and probable polycystic ovary syndrome (PCOS) among parous women were assessed in this study. A total of 557 women were recruited from multi-specialty clinics in eastern Massachusetts. The women were categorized as being diagnosed with PCOS based on medical records and self-reported clinician-diagnoses. A category of "probable PCOS" was created for women without a diagnosis but with ≥ 2 of the following: ovulatory dysfunction (cycle length < 21 or ≥ 35 days), hyperandrogenism (free testosterone concentration > 75th percentile), or elevated anti-Müllerian hormone (AMH) concentration (> 75th percentile). The remaining participants were placed in the "no PCOS" category, and characteristics were compared across groups. Of the total cohort, 9.7% had diagnosed and 9.2% had probable PCOS. The frequency of irregular cycles was similar for diagnosed and probable PCOS. Free testosterone and AMH levels were higher in women with probable than with diagnosed PCOS. Frequency of irregular cycles and both hormones were higher for the 2 PCOS groups vs the no PCOS group. Obesity prevalence for diagnosed PCOS was twice that of probable PCOS (43.9% vs 19.6%), yet the 2 groups had similar HbA1c and adiponectin values. Women with probable PCOS are leaner but have comparable glycemic traits to those with a formal diagnosis, highlighting the importance of assessing biochemical profiles among women with irregular cycles, even in the absence of overweight/obesity.
Collapse
Affiliation(s)
- Wei Perng
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, United States
| | - Victoria W Fitz
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, United States
| | - Kyle Salmon
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, United States
| | - Marie-France Hivert
- Division of Chronic Disease Research across the Lifecourse (CORaL), Department of Population Medicine, Harvard Medical School/Harvard Pilgrim Healthcare Institute, Boston, MA 02215, United States
- Diabetes Unit, Massachusetts General Hospital, Boston, MA 80045, United States
| | - Maryam Kazemi
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA 02115, United States
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research across the Lifecourse (CORaL), Department of Population Medicine, Harvard Medical School/Harvard Pilgrim Healthcare Institute, Boston, MA 02215, United States
| | - Jan Shifren
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, United States
| | - Emily Oken
- Division of Chronic Disease Research across the Lifecourse (CORaL), Department of Population Medicine, Harvard Medical School/Harvard Pilgrim Healthcare Institute, Boston, MA 02215, United States
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA 02115, United States
| | - Jorge E Chavarro
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA 02115, United States
| |
Collapse
|
4
|
Yang Z, Zhou C, Jin L, Pan J. Identification of CCR7 as a potential biomarker in polycystic ovary syndrome through transcriptome sequencing and integrated bioinformatics. Genomics 2025; 117:110968. [PMID: 39608739 DOI: 10.1016/j.ygeno.2024.110968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/10/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder, yet its mechanisms remain elusive. This study employed transcriptome sequencing on granulosa cells from 5 PCOS women and 5 controls, followed by bioinformatic analyses. We identified 684 mRNAs and 167 lncRNAs with significant differential expression. Gene Ontology and KEGG analyses highlighted enrichment in immune and inflammatory responses among these genes. Through CytoHubba plug-in and three machine learning algorithms, CCR7 was identified as the hub gene of PCOS, further validated through analysis of GSE65746, GSE34526 and a cohort of eighty subjects (40 PCOS and 40 controls). Furthermore, a competing endogenous RNA network targeting CCR7 was constructed. Immune infiltration analysis unveiled a significant decrease in monocyte infiltration in PCOS women, with CCR7 expression positively correlated to naïve B cells. Our findings suggest CCR7 and related molecules play a crucial role in the pathogenesis of PCOS, potentially serving as biomarkers for the disorder.
Collapse
Affiliation(s)
- Zuwei Yang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chengliang Zhou
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Jin
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jiexue Pan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
5
|
Wang M, Huang J, Shi Y, Mprah R, Ding H, Zhang S, Li C. Exploring the efficacy of Wenshentiaojing decoction in PCOS: Network pharmacology and mouse model insights. Bioorg Chem 2025; 154:108089. [PMID: 39742672 DOI: 10.1016/j.bioorg.2024.108089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/11/2024] [Accepted: 12/21/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Wenshentiaojing Decoction (WSTJD), a traditional Chinese herbal prescription, was first recorded in the "Ye Tianshi female department secret recipe for diagnosis and treatment ". It has been proven effective in treating polycystic ovary syndrome (PCOS). However, the active ingredients and molecular mechanism of WSTJD against PCOS remain unclear. AIM OF THE STUDY To explore the therapeutic effect and molecular mechanism of WSTJD against PCOS by using network pharmacology and mouse model. MATERIALS AND METHODS Network pharmacology were used to predict active ingredients, potential targets, and pathways of WSTJD against PCOS. Female mice were injected subcutaneously with DHEA (6 mg/100 g body weight) daily to establish a PCOS model and administered with WSTJD and quercetin to observe its therapeutic effect. Thereafter, mouse phenotypes, indicators related to oxidative stress and ferroptosis, and hub genes were determined. RESULTS We identified 144 potential targets for WSTJD in the treatment of PCOS, which were enriched in immune-related signaling pathways such as reactive oxygen species, TNF and IL-17 signaling pathway. Thirteen hub genes were identified by proteinprotein interaction network (PPI) and algorithmic analysis, all of which were oxidative stress-related genes, and five of which, IL6, PTGS2, HIF1A, MTOR and EGFR, were ferroptosis-related genes. Further analysis revealed that quercetin was a key ingredient for WSTJD and that it had superior binding effects with the hub genes. Moreover, WSTJD and quercetin could significantly depress oxidative stress-related indicators and ferroptosis-related gene expression in PCOS mice. Finally, mouse models showed that the expression of the hub genes were consistent with the analysis results. CONCLUSIONS WSTJD and quercetin alleviated PCOS by suppressing oxidative stress and ferroptosis. Quercetin was the key ingredient for WSTJD against PCOS.
Collapse
Affiliation(s)
- Mingming Wang
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221009, PR China; National Experimental Teaching Demonstration Center for Basic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu Province 221009, PR China.
| | - Jing Huang
- Department of Medical Informatics Engineering, Xuzhou Medical University, Xuzhou, Jiangsu Province 221009, PR China
| | - Yue Shi
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221009, PR China; National Experimental Teaching Demonstration Center for Basic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu Province 221009, PR China
| | - Richard Mprah
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221009, PR China
| | - Huanhuan Ding
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221009, PR China; National Experimental Teaching Demonstration Center for Basic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu Province 221009, PR China
| | - Shanshan Zhang
- School of Biological Science, Jining Medical University, Rizhao, Shandong Province 276826, PR China.
| | - Cui Li
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221009, PR China; National Experimental Teaching Demonstration Center for Basic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu Province 221009, PR China.
| |
Collapse
|
6
|
Niu Y, Wang N, Xu Q. Development of an Endoplasmic Reticulum Stress-Related Diagnostic Signature in Polycystic Ovary Syndrome. Reprod Sci 2025; 32:238-250. [PMID: 38955938 DOI: 10.1007/s43032-024-01619-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 06/03/2024] [Indexed: 07/04/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder in premenopausal women. This investigation was to elucidate the underlying mechanism of endoplasmic reticulum stress (ERS) activation in granulosa cells, which has been implicated in the etiology of PCOS. Differentially expressed genes (DEGs) between PCOS and control groups were integrated with ERS gene lists from databases to identify DE-ERS genes, and functional analyses were performed. Univariate regression analysis and the LASSO method were used to select diagnostic factors, followed by establishing a DE-ERS gene-based diagnostic model. A nomogram model was further generated to predict the risk of PCOS. The correlation between ERS gene expression and immune cell proportion was assessed. A total of 14 DE-ERS genes associated with "protein processing in endoplasmic reticulum", "ferroptosis", and "glycerophospholipid metabolism" were selected as PCOS-related factors. An eight-DE-ERS genes-based diagnostic model was developed and displayed satisfactory performance in the training (Area under curve (AUC) = 0.983) and validation datasets (AUC = 0.802). High risk of PCOS can be accurately predicted, which might contribute to clinical decision-making. Moreover, EDEM1 expression was significantly positively correlated with naive B cell infiltration, while PDIA6 was negatively correlated with neutrophil proportion (P < 0.001). We identified eight novel molecules and developed an ERS gene-based diagnostic model in PCOS, which might provide novel insight for finding biomarkers and treatment methods.
Collapse
Affiliation(s)
- Yanxin Niu
- Department of Obstetrics and Gynaecology, Jinhua People's Hospital, No.267, Danxi East Road, Jinhua, 321000, Zhejiang, P.R. China
| | - Nan Wang
- Department of Obstetrics and Gynaecology, Jinhua People's Hospital, No.267, Danxi East Road, Jinhua, 321000, Zhejiang, P.R. China
| | - Qiulian Xu
- Department of Obstetrics and Gynaecology, Jinhua People's Hospital, No.267, Danxi East Road, Jinhua, 321000, Zhejiang, P.R. China.
| |
Collapse
|
7
|
Park KB, Jun KH. Bariatric surgery for treatment of morbid obesity in adults. Korean J Intern Med 2025; 40:24-39. [PMID: 39778524 PMCID: PMC11725483 DOI: 10.3904/kjim.2024.219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/02/2024] [Accepted: 10/20/2024] [Indexed: 01/11/2025] Open
Abstract
Morbid obesity requires active intervention, with treatment options including lifestyle modification, pharmacotherapy, and surgery. As the prevalence of obesity continues to rise in Korea, it is crucial for specialists and general practitioners to have a comprehensive understanding of obesity and its management. Bariatric surgery is the most effective treatment modality for obesity, leading to significant weight loss and metabolic benefits. It involves surgical alterations of normal anatomical structures to improve overall health. Therefore, selecting the appropriate procedure based on the individual characteristics of patients is crucial. This review highlights the two most commonly performed bariatric procedures worldwide, including in Korea: sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB). Furthermore, it provides a comprehensive overview of the surgical techniques involved in SG and RYGB, addresses potential complications, and presents findings from key studies on the weight loss and metabolic outcomes of these surgeries. Additionally, to support clinical application, the review provides outcome data for these procedures based on studies conducted in Korean populations. In addition to SG and RYGB, this review briefly introduces other surgical and endoscopic options, as well as pharmacological treatments that are currently available or may become viable options in the near future.
Collapse
Affiliation(s)
- Ki Bum Park
- Department of Surgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
- Clinic of Metabolic and Bariatric Surgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Kyong-Hwa Jun
- Department of Surgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
- Clinic of Metabolic and Bariatric Surgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| |
Collapse
|
8
|
Nguo K, McGowan M, Cowan S, Davidson Z, Pirotta S, Dordevic AL, Teede H, Hajishafiee M, Carmichael M, Moran LJ. Exploring the physiological factors relating to energy balance in women with polycystic ovary syndrome: a scoping review. Nutr Rev 2025; 83:160-174. [PMID: 38345350 PMCID: PMC11632379 DOI: 10.1093/nutrit/nuad169] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) occurs in 8%-13% of reproductive-aged women and is associated with reproductive, metabolic, and psychological dysfunction. Overweight and obesity are prevalent and exacerbate the features of PCOS. The aim of this review is to evaluate the extent of evidence examining the physiological factors affecting energy homeostasis, which may impact weight gain, weight loss, and weight maintenance in PCOS, and identify research gaps and recommendations for future research. Literature searches using MEDLINE, EMBASE, PsycInfo, AMED, CINAHL, and Cochrane Central Register of Controlled Trials were conducted up to June 22, 2022. Abstracts, non-English-language articles, and reviews were excluded. A total of n = 78 (n = 55 energy intake and n = 23 energy expenditure) primary research papers were included. Papers with multiple outcomes of interest were counted as separate studies. Energy-intake studies (n = 89) focussed on assessing food, nutrient, or supplements stimuli and were grouped into the outcomes of gastrointestinal appetite hormones (n = 43), adipokines (n = 34), subjective appetite (n = 9), functional brain imaging (n = 3), and neuropeptides (n = 0). Energy-expenditure studies (n = 29) were grouped into total energy expenditure (n = 1), resting energy expenditure (n = 15), meal-induced thermogenesis (n = 3), nutrient oxidation (n = 5), and metabolic flexibility (n = 5). Across both energy-intake and -expenditure papers, 60% of the studies compared outcome responses in women with PCOS with a control group. Results were inconsistent, with 57% reporting no differences and 43% reporting altered responses in PCOS compared with controls, including blunted appetite hormone responses, metabolic inflexibility, and reduced energy expenditure. The authors identified that there is inconsistent, yet preliminary, evidence of possible altered physiological factors, which may impact energy balance and weight management. Further work is needed to act on the identified clinical and research gaps to support women with PCOS and health professionals in informing and achieving realistic weight-management goals for women with PCOS. SYSTEMATIC REVIEW REGISTRATION The protocol was prospectively registered on the Open Science Framework on February 16, 2021 (https://osf.io/9jnsm).
Collapse
Affiliation(s)
- Kay Nguo
- Department of Nutrition, Dietetics and Food, Monash University, Melbourne, Victoria, Australia
| | - Margaret McGowan
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Melbourne, Victoria, Australia
| | - Stephanie Cowan
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Melbourne, Victoria, Australia
| | - Zoe Davidson
- Department of Nutrition, Dietetics and Food, Monash University, Melbourne, Victoria, Australia
| | - Stephanie Pirotta
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Melbourne, Victoria, Australia
| | - Aimee L Dordevic
- Department of Nutrition, Dietetics and Food, Monash University, Melbourne, Victoria, Australia
| | - Helena Teede
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Melbourne, Victoria, Australia
| | - Maryam Hajishafiee
- Thompson Institute, University of the Sunshine Coast, Birtinya, Queensland, Australia
| | - Mikaeli Carmichael
- Allied Health and Human Performance, University of South Australia, Adelaide, South Australia, Australia
| | - Lisa J Moran
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Mallya P, Kalthur G, Sravani AB, Lewis SA. Improving the Dehydroepiandrosterone Induced PCOS Rat Model: Interplay of Age, High Fat Diet, and Treatment Regimen on Reproductive and Metabolic Phenotypes. Reprod Sci 2025; 32:187-199. [PMID: 39567467 PMCID: PMC11729200 DOI: 10.1007/s43032-024-01742-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/26/2024] [Indexed: 11/22/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a ubiquitous reproductive condition with triggering hallmarks such as glucose intolerance, hyperandrogenism, and dyslipidemia. Despite the existence of various PCOS animal models, an ideal model which could encompass all PCOS-specific phenotype is of dire need. Dehydroepiandrosterone (DHEA) induced PCOS rats are frequently employed; though, determining the superior model among pubertal and prepubertal rats, incorporation of high fat diet (HFD), and their sustainability remains uncertain. This study aims to examine the age factor, impact of HFD, and DHEA regimen in model development. Prepubertal and pubertal Sprague-Dawley rats were subcutaneously injected with DHEA (6 mg/kg and 60 mg/kg/day, respectively) with and without HFD up to 21 days. Serum testosterone, glucose, lipid profile, ovary morphology, and estrous cycle were evaluated. Following 21 days of treatment with DHEA, pubertal PCOS rats exhibited better reproductive phenotype than prepubertal rats. However, there was no significant difference in the lipid profile. Accordingly, both the age-group rats were concomitantly treated with DHEA and HFD for additional 3 weeks on alternate day basis after model development. The persistence of reproductive and metabolic features on treatment withdrawal were also simultaneously investigated by alienating the rats into continuous and stop dosing groups. The DHEA + HFD and DHEA treated pubertal rats in continuous dosing group showed significant PCOS features (p < 0.05) compared to stop dosing, prepubertal, and control groups. To conclude, continual dosing with DHEA on alternate days for 3 weeks is necessary to sustain metabolic and reproductive phenotypes of PCOS.
Collapse
Affiliation(s)
- Pooja Mallya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Guruprasad Kalthur
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Anne Boyina Sravani
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Shaila A Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
10
|
Wang F, Liu X, Hao X, Wang J, Liu J, Bai C. Oviduct Glycoprotein 1 (OVGP1) Diagnoses Polycystic Ovary Syndrome (PCOS) Based on Machine Learning Algorithms. ACS OMEGA 2024; 9:49054-49063. [PMID: 39713694 PMCID: PMC11656370 DOI: 10.1021/acsomega.4c03111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 11/10/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024]
Abstract
Aims: To investigate the diagnostic value of oviduct glycoprotein 1 (OVGP1) levels for polycystic ovary syndrome (PCOS). Materials and Methods: Serum OVGP1 concentrations were measured by enzyme-linked immunosorbent assay (ELISA). Associations between OVGP1 and endocrine parameters were evaluated by Spearman's correlation analysis. Diagnostic capacity was assessed by utilizing machine learning algorithms and receiver operating characteristic (ROC) curves. Results: OVGP1 levels were significantly decreased in PCOS patients and correlated with the serum follicle-stimulating hormone (FSH) concentration and the luteinizing hormone/follicle-stimulating hormone (LH/FSH) ratio, which are predictors of PCOS occurrence. The diagnostic value of OVGP1 combined with six signatures (LH/FSH, progesterone, total cholesterol, triglyceride, high-density lipoprotein cholesterol, and anti-Müllerian hormone) or three clinical indicators has the potential to significantly improve the accuracy of diagnosing PCOS patients. Conclusion: OVGP1 enhances the ability to diagnose when combined with clinical indicators.
Collapse
Affiliation(s)
| | | | - Xiaoyan Hao
- Department of Clinical Laboratory
Medicine, Xijing Hospital, Fourth Military
Medical University (Air Force Military Medical University), Xi’an 710032, China
| | - Jing Wang
- Department of Clinical Laboratory
Medicine, Xijing Hospital, Fourth Military
Medical University (Air Force Military Medical University), Xi’an 710032, China
| | - Jiayun Liu
- Department of Clinical Laboratory
Medicine, Xijing Hospital, Fourth Military
Medical University (Air Force Military Medical University), Xi’an 710032, China
| | - Congxia Bai
- Department of Clinical Laboratory
Medicine, Xijing Hospital, Fourth Military
Medical University (Air Force Military Medical University), Xi’an 710032, China
| |
Collapse
|
11
|
Guo D, Wang R. Association between visceral adipose surrogates and infertility among reproductive-aged females: a cross-sectional study. Front Endocrinol (Lausanne) 2024; 15:1488309. [PMID: 39726843 PMCID: PMC11669517 DOI: 10.3389/fendo.2024.1488309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background Visceral adipose tissue (VAT) exerts a substantial influence on female infertility. Nevertheless, the relationship between VAT surrogates and female infertility remains ambiguous. Methods This study employed a cross-sectional design and analyzed data from the National Health and Nutrition Examination Survey (NHANES, 2012-2016). Weighted logistic regression models were utilized to examine the association between VAT surrogates and infertility. Furthermore, Receiver Operating Characteristic (ROC) curve analysis was conducted to assess the diagnostic efficacy of these surrogates for infertility. Results Individuals experiencing infertility exhibited markedly elevated levels of the Chinese Visceral Adiposity Index (CVAI) (108.30 vs. 69.86, P<0.001) and Visceral Adiposity Index (VAI) (1.68 vs. 1.35, P<0.001). When considered as a continuous variable, CVAI (odds ratio [OR]: 1.06, 95% confidence interval [CI]: 1.03-1.09, P<0.001), rather than VAI (OR:1.02, 95%CI: 0.98-1.06, P=0.259), demonstrated a significant association with the risk of female infertility. Consistent findings were also evident after dividing participants into 4 subgroups based on CVAI quartiles. Additionally, ROC curves indicated that CVAI exhibited the most robust diagnostic value for female infertility compared to other indices. Subgroup analyses revealed a robust association between CVAI and infertility across different populations. Conclusion Females with elevated CVAI levels faced a significantly heightened risk of infertility in the United States. CVAI holds promise as a valuable tool for stratifying the risk of infertility.
Collapse
Affiliation(s)
- Dongli Guo
- Department of Physiological Obstetrics, Women and Children’s Hospital, Zhumadian Central Hospital, Zhumadian, Henan, China
| | | |
Collapse
|
12
|
Wickham AP, Hewings-Martin Y, Goddard FG, Rodgers AK, Cunningham AC, Prentice C, Wilks O, Kaplan YC, Marhol A, Meczner A, Stsefanovich H, Klepchukova A, Zhaunova L. Exploring Self-Reported Symptoms for Developing and Evaluating Digital Symptom Checkers for Polycystic Ovarian Syndrome, Endometriosis, and Uterine Fibroids: Exploratory Survey Study. JMIR Form Res 2024; 8:e65469. [PMID: 39666967 DOI: 10.2196/65469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/25/2024] [Accepted: 11/13/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Reproductive health conditions such as polycystic ovary syndrome (PCOS), endometriosis, and uterine fibroids pose a significant burden to people who menstruate, health care systems, and economies. Despite clinical guidelines for each condition, prolonged delays in diagnosis are commonplace, resulting in an increase to health care costs and risk of health complications. Symptom checker apps have the potential to significantly reduce time to diagnosis by providing users with health information and tools to better understand their symptoms. OBJECTIVE This study aims to study the prevalence and predictive importance of self-reported symptoms of PCOS, endometriosis, and uterine fibroids, and to explore the efficacy of 3 symptom checkers (developed by Flo Health UK Limited) that use self-reported symptoms when screening for each condition. METHODS Flo's symptom checkers were transcribed into separate web-based surveys for PCOS, endometriosis, and uterine fibroids, asking respondents their diagnostic history for each condition. Participants were aged 18 years or older, female, and living in the United States. Participants either had a confirmed diagnosis (condition-positive) and reported symptoms retrospectively as experienced at the time of diagnosis, or they had not been examined for the condition (condition-negative) and reported their current symptoms as experienced at the time of surveying. Symptom prevalence was calculated for each condition based on the surveys. Least absolute shrinkage and selection operator regression was used to identify key symptoms for predicting each condition. Participants' symptoms were processed by Flo's 3 single-condition symptom checkers, and accuracy was assessed by comparing the symptom checker output with the participant's condition designation. RESULTS A total of 1317 participants were included with 418, 476, and 423 in the PCOS, endometriosis, and uterine fibroids groups, respectively. The most prevalent symptoms for PCOS were fatigue (92%), feeling anxious (87%), BMI over 25 (84%); for endometriosis: very regular lower abdominal pain (89%), fatigue (85%), and referred lower back pain (80%); for uterine fibroids: fatigue (76%), bloating (69%), and changing sanitary protection often (68%). Symptoms of anovulation and amenorrhea (long periods, irregular cycles, and absent periods), and hyperandrogenism (excess hair on chin and abdomen, scalp hair loss, and BMI over 25) were identified as the most predictive symptoms for PCOS, while symptoms related to abdominal pain and the effect pain has on life, bleeding, and fertility complications were among the most predictive symptoms for both endometriosis and uterine fibroids. Symptom checker accuracy was 78%, 73%, and 75% for PCOS, endometriosis, and uterine fibroids, respectively. CONCLUSIONS This exploratory study characterizes self-reported symptomatology and identifies the key predictive symptoms for 3 reproductive conditions. The Flo symptom checkers were evaluated using real, self-reported symptoms and demonstrated high levels of accuracy.
Collapse
|
13
|
Li H, Liu R, Liu J, Qu Y. The Role and Mechanism of Metformin in the Treatment of Nervous System Diseases. Biomolecules 2024; 14:1579. [PMID: 39766286 PMCID: PMC11673726 DOI: 10.3390/biom14121579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Nervous system diseases represent a significant global burden, affecting approximately 16% of the world's population and leading to disability and mortality. These conditions, encompassing both central nervous system (CNS) and peripheral nervous system (PNS) disorders, have substantial social and economic impacts. Metformin, a guanidine derivative derived from a plant source, exhibits therapeutic properties in various health conditions such as cancer, aging, immune-related disorders, polycystic ovary syndrome, cardiovascular ailments, and more. Recent studies highlight metformin's ability to cross the blood-brain barrier, stimulate neurogenesis, and provide beneficial effects in specific neurological disorders through diverse mechanisms. This review discusses the advancements in research on metformin's role and mechanisms in treating neurological disorders within both the central and peripheral nervous systems, aiming to facilitate further investigation, utilization, and clinical application of metformin in neurology.
Collapse
Affiliation(s)
- Hui Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (H.L.); (J.L.)
- Department of General Internal Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China;
| | - Ruhui Liu
- Department of General Internal Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China;
| | - Junyan Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (H.L.); (J.L.)
| | - Yi Qu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (H.L.); (J.L.)
| |
Collapse
|
14
|
Zhou Z, Yang X. An update review of the application of single-cell RNA sequencing in pregnancy-related diseases. Front Endocrinol (Lausanne) 2024; 15:1415173. [PMID: 39717096 PMCID: PMC11663665 DOI: 10.3389/fendo.2024.1415173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Reproductive success hinges on the presence of a robust and functional placenta. Examining the placenta provides insight about the progression of pregnancy and valuable information about the normal developmental trajectory of the fetus. The current limitations of using bulk RNA-sequencing (RNA-seq) analysis stem from the diverse composition of the placenta, hindering a comprehensive description of how distinct trophoblast cell expression patterns contribute to the establishment and sustenance of a successful pregnancy. At present, the transcriptional landscape of intricate tissues increasingly relies on single-cell RNA sequencing (scRNA-seq). A few investigations have utilized scRNA-seq technology to examine the codes governing transcriptome regulation in cells at the maternal-fetal interface. In this review, we explore the fundamental principles of scRNA-seq technology, offering the latest overview of human placental studies utilizing this method across various gestational weeks in both normal pregnancies and pregnancy-related diseases, including recurrent pregnancy loss (RPL), preeclampsia (PE), preterm birth, and gestational diabetes mellitus (GDM). Furthermore, we discuss the limitations and future perspectives of scRNA-seq technology within the realm of reproduction. It seems that scRNA-seq stands out as one of the crucial tools for studying the etiology of pregnancy complications. The future direction of scRNA-seq applications may involve devolving into functional biology, with a primary focus on understanding variations in transcriptional activity among highly specific cell populations. Our goal is to provide obstetricians with an updated understanding of scRNA-seq technology related to pregnancy complications, providing comprehensive understandings to aid in the diagnosis and treatment of these conditions, ultimately improving maternal and fetal prognosis.
Collapse
Affiliation(s)
| | - Xiuhua Yang
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Liu Y, Hocher JG, Ma S, Hu L, Chen H, Zhang X, Gong F, Krämer BK, Lin G, Hocher B. Pre-pregnancy LDL/HDL and total Cholesterol/HDL ratios are strong predictors of gestational diabetes mellitus in women undergoing assisted reproductive technologies. Reprod Biol Endocrinol 2024; 22:155. [PMID: 39639281 PMCID: PMC11619337 DOI: 10.1186/s12958-024-01320-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND & OBJECTIVE To analyze whether there is an association between pre-pregnancy lipid parameters and gestational diabetes mellitus (GDM) in women undergoing assisted reproductive technologies (ART), a group especially at risk for GDM, and if so, which parameter is associated the strongest. METHODS Data was collected at the Reproductive and Genetic Hospital CITIC-Xiangya in Changsha, China from January 2017 to December 2018. The measured lipid parameters include LDL (low-density lipoprotein), HDL (high-density lipoprotein), TC (total cholesterol), and TG (triglycerides). RESULTS 119 (15.5%) of the 767 patients developed GDM. On average, women who developed GDM were older, had a higher BMI, LDL, TC, and TG, and lower HDL. After adjusting for confounders, LDL and HDL showed a significant association with GDM (p < 0.05), but TC and TG did not. Binary LDL/HDL and TC/HDL ratios showed the strongest association with GDM incidence (OR 1.957 [95%CI 1.258-3.044] and 1.942 [1.243-3.034] respectively). Subgroup analysis showed that an elevated LDL/HDL ratio also increased GDM risk in subgroups with a typically lower prevalence of GDM, such as young women with a low BMI and low blood pressure. Both lipid ratios (LDL/HDL and TC/HD) show strong interactions with baseline age, fasting plasma glucose, and LH. CONCLUSIONS In this cohort of Chinese women undergoing ART, pre-pregnancy LDL/HDL and TC/HDL were associated with GDM the strongest from the lipid parameters and could be useful to estimate GDM risk even before ART treatments and pregnancy. CLINICAL TRIAL NUMBER NCT03503006 registered on the 21st of March 2018 (on clinicaltrials.gov). https://clinicaltrials.gov/study/NCT03503006?locStr=Changsha,%20Hunan,%20China&country=China&state=Hunan&city=Changsha&cond=ivf&rank=2 .
Collapse
Affiliation(s)
- Yvonne Liu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, 68167, Mannheim, Germany
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Medical Faculty of Charité Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Johann-Georg Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, 68167, Mannheim, Germany
- Second Faculty of Medicine, Charles University, Prague, 150 06, Czech Republic
| | - Shujuan Ma
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410017, China
| | - Liang Hu
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410017, China
- Key Laboratory of Stem Cells and Reproductive Engineering, Ministry of Health, Changsha, 410017, China
| | - Huijun Chen
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, 68167, Mannheim, Germany
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Medical Faculty of Charité Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Xiaoli Zhang
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, 68167, Mannheim, Germany
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Fei Gong
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410017, China
- Key Laboratory of Stem Cells and Reproductive Engineering, Ministry of Health, Changsha, 410017, China
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, 68167, Mannheim, Germany
- Centre for Preventive Medicine and Digital Health Baden Württemberg (CPDBW), Medical Faculty Mannheim, University of Heidelberg, 68167, Mannheim, Germany
- European Centre for Angioscience ECAS, Medical Faculty Mannheim, University of Heidelberg, 68167, Mannheim, Germany
| | - Ge Lin
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410017, China
- Key Laboratory of Stem Cells and Reproductive Engineering, Ministry of Health, Changsha, 410017, China
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, 68167, Mannheim, Germany.
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China.
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410017, China.
- Institute of Medical Diagnostics, IMD, 12247, Berlin, Germany.
| |
Collapse
|
16
|
Chen Y, Tang YJ, Li X, Wang XM. What can we do for the adolescents with polycystic ovary syndrome? World J Pediatr 2024; 20:1205-1208. [PMID: 39614993 DOI: 10.1007/s12519-024-00857-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 12/12/2024]
Affiliation(s)
- Yao Chen
- Department of Endocrinology, Genetics and Metabolism, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Road, Shanghai 200127, China
| | - Yi-Jun Tang
- Department of Endocrinology, Genetics and Metabolism, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Road, Shanghai 200127, China
| | - Xin Li
- Department of Reproductive Endocrinology, Obstetrics and Gynecology Hospital, School of Medicine, Fudan University, 419 Fangxie Road, Shanghai 200011, China.
| | - Xiu-Min Wang
- Department of Endocrinology, Genetics and Metabolism, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Road, Shanghai 200127, China.
| |
Collapse
|
17
|
Kasa-Vubu JZ, Waisanen A, Sturza J, Padmanabhan V, O’Brien LM. Weight-neutral approach and later sleep midpoint in adolescents with "emerging polycystic ovary syndrome phenotype" as vehicles for sustainable weight loss. F S Rep 2024; 5:402-410. [PMID: 39781077 PMCID: PMC11705593 DOI: 10.1016/j.xfre.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 01/12/2025] Open
Abstract
Objective Incorporate sleep into a novel lifestyle intervention strategy in adolescents with Emerging symptoms of polycystic ovary syndrome (E-PCOS). Design A single-center cohort study. Setting University hospital-based clinic for adolescents with PCOS. Patients Forty-three girls at an age between 10 and 18 years presenting with E-PCOS between March 2015 and September 2017 with clinical signs of androgen excess and/or accelerated weight gain, acanthosis nigricans, irregular periods, or delayed menarche and followed every 6 months for a minimum of 4 visits, to October 2020. Interventions All patients received nutritional counseling, with a goal of "zero weight gain," daily moderate physical activity goals of 45 minutes per day, and education regarding age-appropriate sleep duration. Three treatment strategies for E-PCOS symptoms were applied depending on the chief clinical complaint: anti-insulin approach with metformin; antiandrogen approach with oral contraceptive and spironolactone; and surveillance. Main Outcome Measures Body mass index (BMI) Z-score over time. Alanine Transaminase (ALT) levels as a risk factor for nonalcoholic fatty liver. Results Average number of return visits was 4 with 58% having >4 return visits. Testosterone levels were correlated with ALT (r = 0.68). Weeknight sleep duration was less than age-appropriate recommendations for 63% of participants. Sleep midpoint correlated with ALT levels (r = 0.48). Despite the weight-neutral approach, regression models all demonstrated significant weight loss regardless of menarche status, metformin use, number of visits, and high vs. low ALT groups. Those with the latest sleep midpoint at baseline benefited the most, with BMI Z-score dropping significantly (interaction of time and baseline sleep midpoint from the first visit on school night). Conclusion A novel approach for adolescent girls with E-PCOS that focuses on metabolic endpoints and includes sleep duration and timing as specific targets, led to significant weight loss irrespective of treatment group.
Collapse
Affiliation(s)
- Josephine Z. Kasa-Vubu
- Division of Pediatric Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan
| | - Alexandra Waisanen
- Division of Pediatric Endocrinology, Department of Pediatrics, Nutrition Services, Michigan Medicine, Ann Arbor, Michigan
| | - Julie Sturza
- Department of Pediatrics Biostatistics and Data Management Unit, Michigan Medicine, Ann Arbor, Michigan
| | - Vasantha Padmanabhan
- Division of Pediatric Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan
| | - Louise M. O’Brien
- Departments of Neurology, Division of Sleep Medicine, and Obstetrics & Gynecology, Michigan Medicine, Ann Arbor, Michigan
| |
Collapse
|
18
|
Xiong W, Han L, Tang X, Li R, Chen W, Liu X, Nie H, Qin W, Ling L. Maternal Hypertension and Fecundability: A Population-Based Cohort Study. Hypertension 2024; 81:e173-e184. [PMID: 39328072 DOI: 10.1161/hypertensionaha.124.23562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND The association of hypertension and blood pressure control with fecundability among women is not yet elucidated. The purpose of this study was to evaluate the hypothesis that maternal preconception hypertension would be associated with reduced fecundability and that blood pressure control could reduce excess risk. METHODS Using the National Free Preconception Checkup Projects in Guangdong Province, China, 1422 couples whose female partners had been diagnosed with hypertension and 997 703 reference couples whose female partners were without hypertension were included in this prospective cohort study. Fecundability was measured by time to pregnancy (TTP) and infertility (TTP >12 months). RESULTS Compared with women without hypertension, those with controlled hypertension (time ratio, 1.47 [95% CI, 1.24-1.73]) or uncontrolled hypertension (time ratio, 1.59 [95% CI, 1.34-1.90]) were associated with prolonged TTP and increased risk of infertility (relative risk, 1.19 [95% CI, 1.09-1.31]; relative risk, 1.24 [95% CI, 1.14-1.34]). However, using instrumental variable analyses, there was no significant association between blood pressure control and TTP (time ratio, 0.68 [95% CI, 0.34-1.36]; P=0.270) or infertility (relative risk, 0.97 [95% CI, 0.70-1.34]; P=0.849) among women with hypertension. These results were consistent in the propensity score matching and inverse probability of treatment weighting analyses. CONCLUSIONS Maternal hypertension, with or without controlled blood pressure, was independently associated with prolonged TTP and an increased risk of infertility. These findings may provide insights for the implementation of preconception hypertension screening and the design of future trials.
Collapse
Affiliation(s)
- Wenxue Xiong
- Department of Medical Statistics, School of Public Health (W.X., X.T., R.L., W.C., L.L.), Sun Yat-sen University, Guangzhou, China
| | - Lu Han
- National Health Commission Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), China (L.H., X.L., H.N., W.Q.)
| | - Xijia Tang
- Department of Medical Statistics, School of Public Health (W.X., X.T., R.L., W.C., L.L.), Sun Yat-sen University, Guangzhou, China
| | - Rui Li
- Department of Medical Statistics, School of Public Health (W.X., X.T., R.L., W.C., L.L.), Sun Yat-sen University, Guangzhou, China
| | - Wen Chen
- Department of Medical Statistics, School of Public Health (W.X., X.T., R.L., W.C., L.L.), Sun Yat-sen University, Guangzhou, China
| | - Xiaohua Liu
- National Health Commission Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), China (L.H., X.L., H.N., W.Q.)
| | - Hua Nie
- National Health Commission Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), China (L.H., X.L., H.N., W.Q.)
| | - Weibing Qin
- National Health Commission Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), China (L.H., X.L., H.N., W.Q.)
| | - Li Ling
- Department of Medical Statistics, School of Public Health (W.X., X.T., R.L., W.C., L.L.), Sun Yat-sen University, Guangzhou, China
- Clinical Research Design Division, Clinical Research Center, Sun Yat-sen Memorial Hospital (L.L.), Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
19
|
Nong J, Li H, Yang Y, Lu Q, Sun Y, Yin Q, He H. Low serum hepcidin levels in women with polycystic ovary syndrome: evidence from meta-analysis. Gynecol Endocrinol 2024; 40:2375568. [PMID: 38976752 DOI: 10.1080/09513590.2024.2375568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 06/28/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Iron metabolism plays a significant role in the development of metabolic disorders in women with polycystic ovary syndrome (PCOS). Despite the importance of hepcidin, a key iron regulator, current research on serum hepcidin levels in PCOS patients shows conflicting results. METHODS PubMed, Embase, Web of Science, Cochrane Library and the China National Knowledge Infrastructure (CNKI) database were systematically searched from their inception to 9 September 2023. The search aimed to identify studies in English and Chinese that examined hepcidin levels in women with PCOS compared to healthy control subjects. Standardized mean differences (SMDs) with corresponding 95% confidence intervals (95% CIs) were calculated to evaluate the difference in serum hepcidin levels between women with and without PCOS. RESULTS The meta-analysis included a total of 10 eligible studies, which encompassed 499 PCOS patients and 391 control subjects. The pooled analysis revealed a significant reduction in serum hepcidin levels among the PCOS patients compared to the healthy controls (SMD = -3.49, 95% CI: -4.68 to -2.30, p < .05). There was no statistically significant difference in serum hepcidin levels between PCOS patients with a body mass index (BMI) < 25 and those with a BMI ≥ 25 (p > .05). CONCLUSION The serum hepcidin levels of women with PCOS were significantly lower than those of healthy controls, which suggests that serum hepcidin could be a potential biomarker for PCOS.
Collapse
Affiliation(s)
- Jieou Nong
- Department of Clinical Laboratory, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Hua Li
- Department of Clinical Laboratory, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yunfei Yang
- Department of Clinical Laboratory, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Qiujie Lu
- Department of Clinical Laboratory, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yifan Sun
- Department of Clinical Laboratory, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang City People's Hospital, Guigang, Guangxi, China
| | - Qi Yin
- Department of Clinical Laboratory, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Hongying He
- Department of Gynecology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| |
Collapse
|
20
|
Saleh FL, Starkman H, Furness A, Pfeifer SM, Kives S. Polycystic Ovary Syndrome in Adolescents. Obstet Gynecol Clin North Am 2024; 51:679-693. [PMID: 39510738 DOI: 10.1016/j.ogc.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous endocrinologic disorder that is characterized by oligomenorrhea or amenorrhea and signs of hyperandrogenism. The cause of PCOS remains unknown, but the syndrome is associated with insulin resistance that in turn leads to hyperandrogenism. An accurate and prompt diagnosis of PCOS is important to understanding an adolescent's risk. In adolescents, both oligoovulation and hyperandrogenism are needed for the diagnosis of PCOS and ultrasound is not recommended. Lifelong health consequences of PCOS are significant and include obesity, diabetes, metabolic syndrome, and anovulatory infertility. The symptoms of PCOS can be disturbing to an adolescent girl.
Collapse
Affiliation(s)
- Farrah L Saleh
- Division of Reproductive Endocrinology and Infertility, Weill Medical College of Cornell University, New York-Presbyterian Hospital, New York, NY, USA
| | | | | | - Samantha M Pfeifer
- Division of Reproductive Endocrinology and Infertility, Weill Medical College of Cornell University, New York-Presbyterian Hospital, New York, NY, USA
| | - Sari Kives
- Division of Pediatric Gynecology, University of Toronto, Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
21
|
Liu H, Mo L, Tian X, Fan S, Hu J, Zhang L, Yu B. Correlation between anti-müllerian hormone in polycystic ovarian syndrome with metformin: a systematic review and meta-analysis. Gynecol Endocrinol 2024; 40:2330655. [PMID: 38613449 DOI: 10.1080/09513590.2024.2330655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/05/2024] [Indexed: 04/15/2024] Open
Abstract
OBJECTIVE This study aims to examine the short-term effects of oral metformin (MET) on serum anti-müllerian hormone (AMH) levels and to verify its impact on AMH concentrations in women with polycystic ovary syndrome (PCOS). METHODS The literature search, extending from January 2000 to April 2023, was conducted using databases such as PubMed, Embase, and the Cochrane Central, resulting in the inclusion of 20 studies. These selected studies, evaluated for quality using the Newcastle-Ottawa Scale, investigated changes in AMH levels before and after treatment, with durations ranging from less than three months to over six months. The reported outcomes were quantified as standardized mean differences (SMD) with 95% confidence intervals (CI). This comprehensive systematic review and meta-analysis was registered with the International Prospective Register of Systematic Reviews (PROSPERO) under the registration number CRD42023420705. The statistical analyses were performed using Review Manager 5.4.1. RESULTS ① The study incorporated 20 articles, consisting of 12 prospective studies, 7 randomized controlled trials (RCT), and 1 cross-sectional study. ② Serum AMH levels in patients with PCOS diminish subsequent to the oral administration of MET. ③ Across the spectrum of studies analyzed, a pronounced degree of heterogeneity is evident, potentially ascribed to differential parameters including body mass index (BMI), daily pharmacological dosages, the temporal extent of treatment regimens, criteria of PCOS, and detection Methods. ④ The impact of MET on AMH levels exhibits a dose-responsive trend, with escalating doses of MET being associated with progressively greater declines in AMH concentrations in the patient population. ⑤ For women with PCOS receiving MET therapy, a minimum treatment duration of three months may be necessary to observe a reduction in serum AMH levels. CONCLUSIONS The results of this meta-analysis indicate that MET treatment exerts a suppressive effect on serum AMH levels in women with PCOS. It appears that a treatment duration of at least three months is required to achieve a significant decrease in AMH concentrations. Furthermore, the influence of MET on AMH is dose-dependent, with higher doses correlating with more pronounced reductions in AMH levels among the patients studied.
Collapse
Affiliation(s)
- Hongcen Liu
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen City, Guangdong Province, China
| | - Li Mo
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen City, Guangdong Province, China
| | - Xiaofang Tian
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen City, Guangdong Province, China
| | - Shizhen Fan
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen City, Guangdong Province, China
| | - Jiayi Hu
- The school of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province, China
| | - Lin Zhang
- The school of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province, China
| | - Bohai Yu
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen City, Guangdong Province, China
| |
Collapse
|
22
|
Tong X, Hu Z, Zhou H, Zhang Y, Zhang YL, Zhang S, Jin J. Testosterone-Induced H3K27 Deacetylation Participates in Granulosa Cell Proliferation Suppression and Pathogenesis of Polycystic Ovary Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2326-2340. [PMID: 39243944 DOI: 10.1016/j.ajpath.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/19/2024] [Accepted: 08/16/2024] [Indexed: 09/09/2024]
Abstract
Polycystic ovary syndrome (PCOS) is the leading cause of infertility in reproductive-age women. Hyperandrogenism, polycystic ovaries, and chronic anovulation are its typical clinical features. However, the correlation between hyperandrogenism and ovarian follicle growth aberrations remains poorly understood. To advance our understanding of the molecular alterations in ovarian granulosa cells (GCs) with excessive androgen, epigenetic changes and affected gene expression in human granulosa-lutein cells and immortalized human GCs were evaluated. A PCOS mouse model induced by dihydrotestosterone was also established. This study found that excessive testosterone significantly decreased the acetylation of lysine 27 on histone H3 (H3K27Ac). H3K27Ac chromatin immunoprecipitation-sequencing data showed down-regulated expression of cell cycle-related genes CCND1, CCND3, and PCNA, which was confirmed by real-time quantitative PCR and Western blot analysis. Testosterone application impeding cell proliferation was also shown by Ki-67 immunofluorescence and flow-cytometric analysis. Moreover, testosterone influenced casein kinase 2 alpha (CK2α) nuclear translocation, which increased the phosphorylation level of histone deacetylase 2 (HDAC2). Inhibition of CK2α nuclear translocation or silenced HDAC2 expression efficiently retarded H3K27 acetylation. PCOS mouse model experiments also demonstrated decreased H3K27Ac and enhanced HDAC2 phosphorylation in GCs. Cell proliferation-related genes were also down-regulated in PCOS mouse GCs. In conclusion, hyperandrogenism in human and mouse GCs caused H3K27Ac aberrations, which are associated with CK2α nuclear translocation and HDAC2 phosphorylation, participating in abnormal follicle development in patients with PCOS.
Collapse
Affiliation(s)
- Xiaomei Tong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Zhanhong Hu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Hanjing Zhou
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Yingyi Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Yin-Li Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Jiamin Jin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China.
| |
Collapse
|
23
|
Ryall S, Ohrling H, Stellingwerff T, Black S, Reilly K, Thornton JS. Contraception Choice for Female Endurance Athletes: What's Sport Got to Do With It? A Cross-Sectional Survey. Sports Med 2024; 54:3181-3197. [PMID: 39217588 PMCID: PMC11608295 DOI: 10.1007/s40279-024-02078-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND While there are several prescribed contraceptive methods available, limited evidence exists to guide contraceptive decision-making in the context of endurance sport. OBJECTIVES Study objectives were to characterize current and previous use, perceived impacts, and the decision process around contraceptives in endurance athletes. METHODS This was an online survey study with female endurance athletes recruited through social media and emails to university/club coaches and sport organizations. Quantitative and qualitative data were analyzed with descriptive statistics and conventional content analysis, respectively. RESULTS A total of 323 female endurance athletes participated. Among current contraception users (n = 182), 51% used hormonal intrauterine devices (hIUDs), 29% oral contraceptive pills (OCPs), and 13% nonhormonal IUDs (nhIUDs). hIUD users had the highest perceived positive training (39%) and competition (29%) impacts, citing reduced menstrual bleeding and symptoms as positive side effects. OCP and nhIUD users had higher rates of perceived negative training impacts (OCPs 10%, nhIUDs 30%). For OCP users, 31% reported perceived adverse body composition outcomes and 37% reported negative mood changes. Among nhIUD users, 74% experienced heavier, more irregular menstrual bleeding. Over half of participants were unsure about the impact of their current method on performance. For contraceptive selection, 95% felt that information from physicians was important, yet 32% felt performance was inadequately considered during counseling discussion. Athletes reported less frustration with their contraception choice when counseled in the context of sport. CONCLUSIONS This exploratory study quantifies and qualifies the lived experiences of female endurance athletes with contraception. hIUDs were the most currently used and well-tolerated contraceptives among participants. This research offers valuable insights for athletes seeking contraception and looking to optimize both performance and health, along with the healthcare professionals guiding them.
Collapse
Affiliation(s)
- Stephanie Ryall
- Return to Health and Performance Lab, Department of Family Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada.
| | - Heidi Ohrling
- Return to Health and Performance Lab, Department of Family Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | | | - Stephanie Black
- Department of Obstetrics & Gynecology, Western University, London, ON, Canada
| | - Kristen Reilly
- Return to Health and Performance Lab, Department of Family Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Jane S Thornton
- Department of Family Medicine, Western University, London, ON, Canada
| |
Collapse
|
24
|
Wu HH, Zhu Q, Liang N, Xiang Y, Xu TY, Huang ZC, Cai JY, Weng LL, Ge HS. CISD2 regulates oxidative stress and mitophagy to maintain the balance of the follicular microenvironment in PCOS. Redox Rep 2024; 29:2377870. [PMID: 39010730 PMCID: PMC467114 DOI: 10.1080/13510002.2024.2377870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024] Open
Abstract
OBJECTIVES To observe the CISD2 expression among PCOS patients and to explore its profound impact on the follicular microenvironment. Moreover, we want to elucidate the intricate mechanistic contribution of CISD2 to the onset and progression of PCOS. METHODS Oxidase NOX2, mitophagy-related proteins, and CISD2 were detected by WB. The changes in mitochondrial structure and quantity were observed by transmission electron microscopy. Mitochondrial and lysosome colocalization was used to detect the changes of mitophagy. MDA kit, GSH and GSSG Assay kit and ROS probe were used to detect oxidative stress damage. RESULTS We found that CISD2, mitophagy and oxidase in the GCs of PCOS patients were significantly increased. Testosterone stimulation leads to the increase of oxidase, mitophagy, and CISD2 in KGN cells. CISD2 inhibition promoted the increase of mitophagy, and the activation of mitochondria-lysosome binding, while alleviating the oxidative stress. CONCLUSIONS Inhibition of CISD2 can improve the occurrence of oxidative stress by increasing the level of mitophagy, thus affecting the occurrence and development of PCOS diseases.
Collapse
Affiliation(s)
- Hong-Hui Wu
- Graduate School, Dalian Medical University, Liaoning, People’s Republic of China
- Reproduction Medicine Centre, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, People’s Republic of China
| | - Qi Zhu
- Reproduction Medicine Centre, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, People’s Republic of China
- Graduate School, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Na Liang
- Graduate School, Dalian Medical University, Liaoning, People’s Republic of China
- Reproduction Medicine Centre, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, People’s Republic of China
| | - Yu Xiang
- Reproduction Medicine Centre, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, People’s Republic of China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Tian-Yue Xu
- Reproduction Medicine Centre, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, People’s Republic of China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Zi-Chao Huang
- Reproduction Medicine Centre, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, People’s Republic of China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Jie-Yu Cai
- Reproduction Medicine Centre, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, People’s Republic of China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Ling-Lin Weng
- Reproduction Medicine Centre, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, People’s Republic of China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Hong-Shan Ge
- Graduate School, Dalian Medical University, Liaoning, People’s Republic of China
- Reproduction Medicine Centre, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, People’s Republic of China
- Graduate School, Nanjing Medical University, Nanjing, People’s Republic of China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
25
|
Guo B, Shen Y, Dai Z, Yimamu K, Sun J, Pei L. A nomogram to predict the risk of insulin resistance in Chinese women with polycystic ovary syndrome. Front Endocrinol (Lausanne) 2024; 15:1446827. [PMID: 39665024 PMCID: PMC11631621 DOI: 10.3389/fendo.2024.1446827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
Background Insulin resistance (IR) is considered a major driver of the pathophysiology of polycystic ovary syndrome (PCOS), mediating the progression of hyperandrogenism and metabolic and reproductive dysfunction in patients with PCOS. Early detection of the risk of concurrent IR is essential for women with PCOS. To address this need, this study developed a predictive nomogram for assessing the risk of IR in women with PCOS, aiming to provide a tool for risk stratification and assist in clinical decision-making. Methods Patients with untreated PCOS-IR diagnosed in a single-center retrospective cohort study from January 2023 to December 2023 were included for nomogram construction and validation. The area under the ROC curve (AUC), calibration curve, Hosmer-Lemeshow (H-L) goodness-of-fit test, and decision curve analysis (DCA) were used to evaluate the nomogram's discrimination, calibration, and clinical decision performance. A risk stratification model based on the nomogram was then developed. Results A total of 571 patients were included in the study; 400 patients enrolled before September 2023 were divided into the training and validation sets, and 171 patients enrolled later were used as the external validation set. The variables identified by logistic regression and the random forest algorithm-body mass index (BMI, OR 1.43), triglycerides (TG, OR 1.22), alanine aminotransferase (ALT, OR 1.03), and fasting plasma glucose (FPG, OR 5.19)-were used to build the nomogram. In the training, internal validation, and external validation sets, the AUCs were 0.911 (95% CI 0.878-0.911), 0.842 (95% CI 0.771-0.842), and 0.901 (95% CI 0.856-0.901), respectively. The nomogram showed good agreement between predicted and observed outcomes, and patients were categorized into low-, medium-, and high-risk groups based on their scores. Conclusions Independent predictors of untreated PCOS-IR risk were incorporated into a nomogram that effectively classifies patients into risk groups, providing a practical tool for guiding clinical management and early intervention.
Collapse
Affiliation(s)
| | | | | | | | - Jianhua Sun
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Lixia Pei
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
26
|
Zhang H, Qiu W, Zhou P, Shi L, Chen Z, Yang Y, Lu Y, Zhou L, Zhang H, Cheng M, Ye Y, Li R. Obesity is associated with SHBG levels rather than blood lipid profiles in PCOS patients with insulin resistance. BMC Endocr Disord 2024; 24:254. [PMID: 39587600 PMCID: PMC11587586 DOI: 10.1186/s12902-024-01789-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder characterized by hormonal imbalances and insulin resistance (IR). Among the metabolic abnormalities associated with PCOS, obesity is often present concurrently. Nevertheless, the correlation between obesity, sex hormone levels, and blood lipid profiles in PCOS patients with IR remains uncertain. METHODS This is a cross-sectional study including a total of 206 Chinese women diagnosed with PCOS, enrolled between March 2016 and December 2021. The participants' anthropometric measurements, such as weight, height, waist circumference, and hip circumference, were recorded. Additionally, fasting blood samples were collected to measure various parameters, including fasting glucose, insulin levels, lipid profiles, and sex hormone levels. RESULTS Our findings highlight that obesity exhibited a significant correlation with lower levels of sex hormone binding globulin (SHBG) and elevated levels of free androgen index (FAI), fasting insulin, and HOMA-IR in PCOS patients diagnosed with IR. However, no significant association between obesity and blood lipid profiles was observed within this particular group of women. CONCLUSION This study suggests that among PCOS patients with IR, lower levels of SHBG and higher levels of FAI are associated with obesity. These findings indicate that SHBG and FAI may have the potential to serve as a biomarker for the initial identification and prognosis of IR in PCOS patients. TRIAL REGISTRATION Retrospectively registered on 25/04/2020 at ClinicalTrials.gov Identifer: NCT04264832.
Collapse
Affiliation(s)
- Haolin Zhang
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
| | - Weiyu Qiu
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ping Zhou
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
| | - Li Shi
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
| | - Ziting Chen
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Yang Yang
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Yonghao Lu
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Lifei Zhou
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Hua Zhang
- Research Centre of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Ming Cheng
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
| | - Yang Ye
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China.
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
27
|
Long JR, Parker M, Jumani S, Ahmed A, Huynh V, Gomez-Lobo V. Effect of Lifestyle Modifications on Polycystic Ovary Syndrome in Predominantly Young Adults: A Systematic Review. J Pediatr Adolesc Gynecol 2024:S1083-3188(24)00342-5. [PMID: 39577757 DOI: 10.1016/j.jpag.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 10/25/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVE To improve counseling and outcomes for the adolescent population (ages 10-24-years-old), with polycystic ovary syndrome we conducted a systematic review of randomized controlled trials with the primary objective to generate evidence-based recommendations for which lifestyle interventions with or without medications lead to the best outcomes. METHODS A literature search was conducted. Randomized controlled trials on lifestyle interventions with or without medications in the adolescent population were included. Nonrandomized trials, case-control studies, observational studies, and animal studies were excluded. Of 3,699 articles, 13 studies including 789 participants were included. Each included study was assessed for bias using the Cochrane Risk of Bias 2 tool. Due to significant interstudy heterogeneity, meta-analysis was infeasible; we synthesized results across lifestyle intervention/control types and outcome. RESULTS Thirteen studies met inclusion criteria. These studies offer mixed support for lifestyle interventions improving hyperandrogenism. There is some evidence that lifestyle interventions improve menstrual regularity, cardiometabolic health, and metabolic function. Almost all studies found reduced body mass index, adiposity among participants who completed combined exercise and diet, exercise only, and diet only interventions. CONCLUSIONS The studies in this systematic review demonstrated that lifestyle interventions incorporating increased physical activity and/or healthy dietary choices show beneficial effects in the adolescent population aged ≥ 18-years-old with polycystic ovary syndrome. Medications may also play a key role in treating the disorder. More quality research is needed to identify specific lifestyle interventions that optimize the management of polycystic ovary syndrome amongst those aged 10-17-years-old as well.
Collapse
Affiliation(s)
- Jessica R Long
- University of Chicago Obstetrics & Gynecology Department, Chicago, IL.
| | | | - Sanjay Jumani
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, MD
| | - Aisha Ahmed
- Howard University School of Medicine Obstetrics & Gynecology Department, Washington, DC
| | - Victoria Huynh
- Baylor College of Medicine Obstetrics & Gynecology Department, Houston, TX
| | - Veronica Gomez-Lobo
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, MD
| |
Collapse
|
28
|
Li J, Fan L, Wei J, Huang W. Elucidating the pathophysiology of polycystic ovary syndrome: Construction and analysis of a ceRNA network in cumulus cells. Reprod Biol 2024; 25:100916. [PMID: 39566253 DOI: 10.1016/j.repbio.2024.100916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/13/2024] [Accepted: 06/16/2024] [Indexed: 11/22/2024]
Abstract
Polycystic Ovary Syndrome (PCOS) is a complex endocrine disorder with elusive molecular mechanisms. This study explores the competitive endogenous RNA (ceRNA) regulatory network in the cumulus cells of PCOS patients. ceRNAs are transcripts like mRNAs, miRNAs, and lncRNAs that competitively bind shared miRNAs, regulating gene expression post-transcriptionally. We analyzed mRNA, microRNA (miRNA), and long non-coding RNA (lncRNA) from two cohorts: 12 PCOS patients and 11 healthy controls (dataset GSE10946), and 5 PCOS patients and 5 healthy controls (dataset GSE72274). These microarray datasets, obtained from the Gene Expression Omnibus (GEO), helped us identify differentially expressed mRNAs, miRNAs, and lncRNAs. Our analysis revealed a significant ceRNA network, which may play a crucial role in the pathophysiology of PCOS. In this network, 5 lncRNAs, 3 miRNAs, and 36 mRNAs were identified as differentially expressed. These elements form a complex regulatory schema influencing key cellular processes related to the disease, such as cell cycle regulation and response to estrogen. The HOXA11-AS-hsa-miR-454-3p-CCND2 network emerged as a potentially valuable biomarker for PCOS diagnosis, supported by Receiver Operating Characteristic (ROC) curve analysis indicating strong predictive power. Our findings suggest that the ceRNA interactions in PCOS cumulus cells provide a deeper understanding of the disease's molecular basis and offer new avenues for therapeutic intervention. This in silico study lays the groundwork for further experimental validation of these ceRNA networks as targets for PCOS treatment.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi 545616, China; Department of Reproductive Medicine, Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi 545001, China; Guangxi Clinical Research Center for Obstetrics and Gynecology, China; Liuzhou Key Laboratory of Gynecologic Tumor, China
| | - Li Fan
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi 545616, China; Department of Reproductive Medicine, Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi 545001, China; Guangxi Clinical Research Center for Obstetrics and Gynecology, China; Liuzhou Key Laboratory of Gynecologic Tumor, China
| | - Jiajia Wei
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi 545616, China; Department of Reproductive Medicine, Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi 545001, China; Guangxi Clinical Research Center for Obstetrics and Gynecology, China; Liuzhou Key Laboratory of Gynecologic Tumor, China
| | - Wenjie Huang
- Department of Reproductive Medicine, Guangzhou Women and Children's Medical center Liuzhou Hospital, Liuzhou, Guangxi 545616, China; Department of Reproductive Medicine, Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi 545001, China; Guangxi Clinical Research Center for Obstetrics and Gynecology, China; Liuzhou Key Laboratory of Gynecologic Tumor, China.
| |
Collapse
|
29
|
Cooney LG, Gyorfi K, Sanneh A, Bui LM, Mousa A, Tay CT, Teede H, Stener-Victorin E, Brennan L. Increased Prevalence of Binge Eating Disorder and Bulimia Nervosa in Women With Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis. J Clin Endocrinol Metab 2024; 109:3293-3305. [PMID: 39115340 DOI: 10.1210/clinem/dgae462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Indexed: 11/19/2024]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is associated with disordered eating/eating disorders, but prior meta-analyses are limited by small numbers. OBJECTIVE To inform the 2023 International PCOS Guideline, we performed a systematic review and meta-analysis evaluating the prevalence of disordered eating/eating disorders among women with and without PCOS. METHODS Ovid MEDLINE, EMBASE, PsycInfo, and All EMB were searched from inception through February 1, 2024, for studies that compared prevalences of eating disordered/disordered eating in adolescent or adult women. Random effects meta-analyses were used to estimate the pooled odds ratios (OR) or standardized mean differences (SMD) of outcomes in women with PCOS compared to controls. Methodological quality was assessed by the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) system, and included studies were assessed for risk of bias. RESULTS Of 1352 articles identified, 20 were included, with a total of 28 922 women with PCOS and 258 619 controls. Individuals with PCOS had higher odds of any eating disorder (OR: 1.53 [1.29, 1.82], 8 studies), which persisted in studies where PCOS was diagnosed by Rotterdam criteria (OR: 2.88 [1.55, 5.34], 4 studies). Odds of bulimia nervosa, binge eating disorder, and disordered eating, but not anorexia nervosa, were increased in PCOS. Mean disordered eating scores were higher in PCOS (SMD: 0.52 [0.28, 0.77], 13 studies), including when stratified by normal and higher weight body mass index. Most included studies were of moderate quality, with no evidence of publication bias. CONCLUSION Our study informs the 2023 PCOS Guideline recommendations for consideration of the risk of disordered eating/ eating disorders in care of women with PCOS, regardless of weight, especially during providing lifestyle counseling.
Collapse
Affiliation(s)
- Laura G Cooney
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 54562, USA
| | - Kaley Gyorfi
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 54562, USA
| | - Awa Sanneh
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 54562, USA
| | - Leeann M Bui
- Department of Obstetrics and Gynecology, Santa Clara Valley Medical Center, Santa Clara, CA 95128, USA
| | - Aya Mousa
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, 3800, Australia
| | - Chau Thien Tay
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, 3800, Australia
- Endocrine and Diabetes Units, Monash Health, Cayton, 3168, Australia
| | - Helena Teede
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, 3800, Australia
- Endocrine and Diabetes Units, Monash Health, Cayton, 3168, Australia
| | | | - Leah Brennan
- School of Psychology and Public Health, La Trobe University, Melbourne, 3086, Australia
| |
Collapse
|
30
|
Martinez Guevara D, Vidal Cañas S, Palacios I, Gómez A, Estrada M, Gallego J, Liscano Y. Effectiveness of Probiotics, Prebiotics, and Synbiotics in Managing Insulin Resistance and Hormonal Imbalance in Women with Polycystic Ovary Syndrome (PCOS): A Systematic Review of Randomized Clinical Trials. Nutrients 2024; 16:3916. [PMID: 39599701 PMCID: PMC11597640 DOI: 10.3390/nu16223916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Polycystic ovary syndrome is a common endocrine disorder in women of reproductive age characterized by insulin resistance and hormonal imbalances. Recent research suggests that probiotics and synbiotics may improve these parameters by modulating the gut microbiota. This study systematically reviewed randomized clinical trials evaluating the impact of probiotic, prebiotic, and synbiotic supplementation on insulin resistance and hormonal parameters in women with PCOS. Methods: Exhaustive searches were conducted in PubMed, Cochrane CENTRAL, Scopus, Web of Science, and Embase, following PRISMA guidelines. Randomized trials assessing supplementation with probiotics, prebiotics, or synbiotics for at least 8 weeks in women diagnosed with PCOS according to the Rotterdam criteria were included. Data on participants, interventions, and outcomes related to insulin resistance and hormones were extracted. Results: Eleven studies from Iran involving overweight or obese women aged 15 to 48 were included. Probiotic and synbiotic supplementation showed significant improvements in insulin resistance (reductions in HOMA-IR, fasting glucose, and insulin), lipid profiles (decreased LDL and triglycerides; increased HDL), and hormonal balance (increased SHBG, decreased total testosterone). Synbiotics had more pronounced effects than probiotics or prebiotics alone. Adherence was high, and side effects were minimal. Conclusions: Despite promising results, limitations such as small sample sizes, homogeneous populations, and short intervention durations limit the generalization of the findings. Larger, longer, multicenter trials with diverse populations and standardized methodologies are needed to confirm the efficacy and safety of synbiotics in managing PCOS. Integrating these interventions could improve clinical management and quality of life for affected women, but additional evidence is required to support widespread use.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yamil Liscano
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 5183000, Colombia; (D.M.G.); (S.V.C.); (I.P.); (A.G.); (M.E.); (J.G.)
| |
Collapse
|
31
|
Fu X, Cao W, Ye F, Bei J, Du Y, Wang L. Astaxanthin compound nutrient improved insulin resistance, hormone levels, embryo quality and pregnancy outcomes in polycystic ovary syndrome patients undergoing in vitro fertilization/intracytoplasmic sperm injection. Drug Discov Ther 2024; 18:296-302. [PMID: 39428502 DOI: 10.5582/ddt.2024.01036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
This study aimed to evaluate the effect of astaxanthin compound nutrient (ACN) complementary therapy on pregnancy outcomes in polycystic ovary syndrome (PCOS) patients undergoing in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI). This study enrolled 92 patients with PCOS who were continuously supplemented with ACN for three months prior to IVF/ICSI treatment from 2021 to 2023, and selected 92 patients who did not receive the treatment during the same period as controls. Baseline characteristics, ovulation induction outcomes, and pregnancy outcomes were compared between the two groups. In addition, the body mass index (BMI), anti-Müllerian hormone (AMH), antral follicle counting (AFC), fasting blood glucose (FBG), fasting insulin (FINS), homeostasis model assessment of insulin resistant (HOME-IR), and basal sex hormones of the supplementary group patients before and after treatment were compared. The results showed that there were no significant differences in the patient's duration of stimulation, total gonadotropin dose, peak E2 levels, and number of retrieved oocytes between the two groups. However, the number of 2 pronucleus (PN) fertilization, transferable embryos, and high-quality embryos was significantly higher in the ACN group compared with the control group. For both fresh and frozen embryo transplantation, positive pregnancy outcomes increased in PCOS patients who received supplementation of ACN for 3 months. In addition, after 3 months of supplementing with ACN, the patient's BMI, AMH, fasting insulin, HOME-IR, basal luteinising hormone (bLH), and basal testosterone (bT) decreased compared to before treatment. This study suggested that ACN improved insulin resistance, hormone levels, embryo quality and pregnancy outcomes in PCOS patients.
Collapse
Affiliation(s)
- Xiayan Fu
- Reproductive Medicine Center, Zhoushan Maternal and Child Health Care Hospital, Zhoushan, Zhejiang, China
| | - Wenli Cao
- Reproductive Medicine Center, Zhoushan Maternal and Child Health Care Hospital, Zhoushan, Zhejiang, China
| | - Feijun Ye
- Reproductive Medicine Center, Zhoushan Maternal and Child Health Care Hospital, Zhoushan, Zhejiang, China
| | - Jialu Bei
- Reproductive Medicine Center, Zhoushan Maternal and Child Health Care Hospital, Zhoushan, Zhejiang, China
| | - Yan Du
- Clinical Research Unit, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Ling Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| |
Collapse
|
32
|
Bao X, Yan D, Yang J, Zhang Z, Yuan B. Role of ERβ in the ovary and ovary related diseases. Gene 2024; 927:148678. [PMID: 38906392 DOI: 10.1016/j.gene.2024.148678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 06/01/2024] [Accepted: 06/07/2024] [Indexed: 06/23/2024]
Abstract
Estrogen and estrogen receptors (ERα and ERβ) regulate a multitude of complicated physiological and pathological processes. Jan-Ake Gustafsson's group discovered ERβ in 1996, this crucial finding gives us new insights into the understanding of estrogen signaling. ERβ is highly expressed in the ovary and particularly exists in granulosa cells (GCs). ERβ is a key transcription factor in the maintenance of ovarian granulosa cell growth, differentiation, and homeostasis, and the ovulation function of ovarian follicles and oocytes. Additionally, ERβ can modulate the steroidogenic transcriptional program through phosphorylation and regulate both gonadotropin response and FOXL2 expression within the ovary. In this review, we focus on the role of ERβ in regulating ovarian granulosa cell development and homeostasis, particularly its significance in ovarian cancer (OC), premature ovarian failure (POF), and polycystic ovary syndrome (PCOS). It also highlights the prospects of small molecule compounds targeting ERβ, providing a new strategy for the treatment of ovarian-related diseases.
Collapse
Affiliation(s)
- Xuewei Bao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, People's Republic of China
| | - Di Yan
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, People's Republic of China; Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui 230032, People's Republic of China
| | - Jing Yang
- Experimental Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, People's Republic of China
| | - Zhen Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, People's Republic of China.
| | - Bin Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, People's Republic of China.
| |
Collapse
|
33
|
Qin Z, Zhang D, Cao G, Li H. Progestin-based pharmacotherapy in fertility preservation in early endometrial cancer. Front Oncol 2024; 14:1487008. [PMID: 39588311 PMCID: PMC11586232 DOI: 10.3389/fonc.2024.1487008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/14/2024] [Indexed: 11/27/2024] Open
Abstract
Endometrial cancer is a common tumor of the female reproductive system. In recent years, as the age of onset of the disease has gradually become younger, this has caused distress to some young patients with reproductive needs, and the active search for methods of preserving reproductive function has gradually attracted attention. In this paper, we will systematize the current status of progestin-based pharmacotherapy in combination with other drug therapies in the conservative management of early-stage endometrial cancer. With the expectation of providing a reference for the treatment of early stage endometrial cancer patients in China and for the in-depth development of related research in this field.
Collapse
Affiliation(s)
| | | | | | - Hua Li
- Beijing Chaoyang Hospital, Capital Medical University,
Beijing, China
| |
Collapse
|
34
|
Zhang Y, Martin L, Mustieles V, Ghaly M, Archer M, Sun Y, Torres N, Coburn-Sanderson A, Souter I, Petrozza JC, Botelho JC, Calafat AM, Wang YX, Messerlian C. Per- and polyfluoroalkyl substances exposure is associated with polycystic ovary syndrome risk among women attending a fertility clinic. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175313. [PMID: 39117221 PMCID: PMC11357523 DOI: 10.1016/j.scitotenv.2024.175313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/26/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Previous studies reported that exposures to per- and polyfluoroalkyl substances (PFAS), largely in higher exposed populations, were associated with elevated risk of polycystic ovary syndrome (PCOS). However, studies evaluating PCOS risk in populations with lower background exposures to PFAS are limited. This study aimed to examine the associations between serum PFAS concentrations and PCOS risk among women attending a U.S. academic fertility clinic during 2005-2019. A total of 502 females who sought fertility evaluation and assisted reproduction treatments were included. Nine PFAS were quantified in non-fasting serum samples collected at study entry. Diagnosis of PCOS was based on the Rotterdam criteria. We used logistic regression to examine the odds ratio (OR) of PCOS in relation to individual PFAS concentrations (continuous and by tertiles) and quantile g-computation (QGC) and Bayesian Kernel Machine Regression (BKMR) to examine the joint associations of PFAS mixture with PCOS. Most participants were White and had a graduate degree or higher. Per doubling of serum perfluorooctane sulfonate (PFOS) and perfluorohexane sulfonate (PFHxS) concentrations were associated with higher odds of PCOS [OR (95%CI): 1.70 (1.06, 2.81) and 1.45 (1.02, 2.08) for PFOS and PFHxS respectively]. There was a dose-response relationship of PFOS with PCOS risk (p of trend by PFOS tertiles = 0.07). Both QGC and BKMR identified PFOS as the most important contributor among the mixture to PCOS risk. No clear joint effects were found for other PFAS or PFAS mixtures on PCOS risk. Our findings are consistent with existing evidence in populations with higher background PFAS concentrations and highlight the adverse effects of PFAS exposure on reproductive health. Findings can inform public health measures and clinical care to protect populations vulnerable to PCOS, in part, due to environmental exposures.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Leah Martin
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Vicente Mustieles
- Instituto de Investigación Biosanitaria Ibs GRANADA, Spain; University of Granada, Center for Biomedical Research (CIBM), Spain.; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Spain
| | - Mina Ghaly
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Madeleine Archer
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yang Sun
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Nicole Torres
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ayanna Coburn-Sanderson
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Irene Souter
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital Fertility Center, Boston, MA, USA
| | - John C Petrozza
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital Fertility Center, Boston, MA, USA
| | - Julianne C Botelho
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Antonia M Calafat
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Yi-Xin Wang
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Carmen Messerlian
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital Fertility Center, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
35
|
Yin S, Zhu F, Zhou Q, Chen M, Wang X, Chen Q. Lack of Efficacy of Pomegranate Supplementation on Insulin Resistance and Sensitivity: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Phytother Res 2024. [PMID: 39499092 DOI: 10.1002/ptr.8362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/20/2024] [Accepted: 09/01/2024] [Indexed: 11/07/2024]
Abstract
The objective of this study is to assess the impact of pomegranate supplements on insulin resistance (IR) and insulin sensitivity through a systematic review and meta-analysis of randomized controlled trials (RCTs). Additionally, we aim to analyze the differences in efficacy among various pomegranate extracts and the sensitivity of different diseases to pomegranate supplementation. We conducted searches in PubMed, Embase, Web of Science, and Cochrane Library up to October 30, 2023, for relevant studies published in English. The treatment group required the intake of pomegranate extract for a minimum of 4 weeks, with no restrictions on the extract type. The control group received a placebo or a treatment excluding pomegranate extract. The primary outcome was homeostatic model assessment for insulin resistance (HOMA-IR) and fasting insulin (FI), and the secondary outcome was quantitative insulin sensitivity check index (QUICKI). RoB 2 was used to assess the risk of bias in the original studies. We pre-specified subgroup analyses based on types of intervention, intervention duration, health condition, and intervention dose. Sensitivity analysis was conducted to validate result stability, utilizing Begg's test and Egger's test for publication bias. Data synthesis and analysis were performed using Stata 15.1 software. This study included a total of 15 RCTs with 673 participants conducted in 7 countries. Risk of bias results indicated an overall low risk of bias of the articles. Participants included healthy individuals, overweight and obese individuals, non-alcoholic fatty liver disease (NAFLD) patients, type 2 diabetes (T2DM) patients, polycystic ovary syndrome (PCOS) patients, metabolic syndrome (MS) patients, and individuals with hyperlipidemia. Pomegranate extract variations included pomegranate juice (PJ), pomegranate seed oil (PSO) capsule, pomegranate/pomegranate peel (PP) extract capsule, and pomegranate peel-added bread. The control groups primarily received placebo treatments with varying dosage and frequency. No adverse reactions were reported in any of the studies. The summary results showed that compared to the control groups, pomegranate extract had no significant impact on improving HOMA-IR levels in participants (WMD = -0.03, 95%CI: -0.37 to 0.31, and p = 0.851) and FI (WMD = -0.03, 95%CI: -0.42 to 0.36, and p = 0.862). Additionally, there was no significant advantage of pomegranate extract on QUICKI changes in T2DM and PCOS patients (WMD = 0.00, 95%CI: 0.00 to 0.01, and p = 0.002). Subgroup analysis results indicated that pomegranate extract could improve HOMA-IR levels in PCOS patients (WMD = -0.42, 95%CI: -0.54 to -0.29, and p < 0.001) and FI levels in T2DM, PCOS, and NAFLD patients. Our results indicate that pomegranate extract only improves HOMA-IR and FI levels in PCOS patients and FI levels in T2DM and NAFLD patients. No significant difference has been found for HOMA-IR, FI, or QUICKI in other metabolic diseases. The current evidence suggests that we should interpret the value of pomegranate extract in regulating IR and sensitivity cautiously. In the future, there is a need for more rigorously designed RCTs to specifically evaluate the impact of pomegranate supplementation on insulin sensitivity in patients with NAFLD, PCOS, and T2DM.
Collapse
Affiliation(s)
- Shao Yin
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Fengya Zhu
- Traditional Chinese Medicine Department, Zigong First People's Hospital, Zigong, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Zhou
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Miao Chen
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Xia Wang
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Qiu Chen
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
36
|
Goparaju P, Gragnoli C. Implication of vasopressin receptor genes (AVPR1A and AVPR1B) in the susceptibility to polycystic ovary syndrome. J Ovarian Res 2024; 17:214. [PMID: 39501331 PMCID: PMC11536872 DOI: 10.1186/s13048-024-01515-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 09/13/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a complex heterogenous disorder manifesting with various reproductive, endocrine, and metabolic derangements such as insulin resistance and hyperglycemia. The arginine vasopressin peptide (AVP), also called or antidiuretic hormone (ADH), modulates metabolic functions such as glucose hemostasis, insulin sensitivity, and lipid metabolism via binding to two central and peripheral receptors (AVPR1A and AVPR1B). In the present study, we aimed to detect whether the AVPR1A and AVPR1B genes confer risk for PCOS. METHODS In peninsular Italian families, we tested 7 variants in the AVPR1B gene and 2 variants in the AVPR1A gene via Pseudomarker for linkage and linkage joint to association (i.e.., linkage disequilibrium) with PCOS. RESULTS We identified two risk variants in each gene, significantly associated with the risk of PCOS. CONCLUSION To the best of our knowledge, this is the first study to report risk variants in AVPR1A and AVPR1B genes in association with PCOS. However, replication in other ethnic groups as well as functional studies are needed to confirm these results.
Collapse
Affiliation(s)
- Pruthvi Goparaju
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE, 68124, USA
| | - Claudia Gragnoli
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE, 68124, USA.
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA.
- Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, 00197, Rome, Italy.
| |
Collapse
|
37
|
Rentería MS, Montoya JAP, Romero GS, de Jesús González Piñuelas L, López-Barradas AM, Granados-Portillo O, Chagollán MG, Suárez ALP, Gillevet PM, Magaña NV, Peña Rodríguez M. Impact of Dietary Patterns and Serum Amino Acid Profile on Metabolic Syndrome Development in Mexican Women with Polycystic Ovary Syndrome. Int J Mol Sci 2024; 25:11821. [PMID: 39519371 PMCID: PMC11547086 DOI: 10.3390/ijms252111821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is the main endocrine disorder in women of reproductive age worldwide. This condition is often associated with various metabolic alterations that contribute to the development of metabolic syndrome (MetS). Recent research suggests that branched-chain amino acid (BCAA) dysregulation is observed in PCOS. This study aims to investigate the relationship between dietary patterns, body composition, metabolic analytes, and serum amino acid levels in Mexican women with PCOS. Utilizing a cross-sectional design, we found that both study groups, PCOS (n = 24) and PCOS + MetS (n = 21), exhibited increased relative fat mass and dietary habits characterized by high simple sugar intake and low protein consumption, correlating with levels of relative fat mass and leptin. Notably, serum concentrations of BCAAs and glutamic acid were significantly elevated in the PCOS + MetS group. Our findings suggest that a metabolic approach may enhance the prediction and management of MetS in women with PCOS, highlighting the importance of dietary interventions in this population.
Collapse
Affiliation(s)
- Midory Sánchez Rentería
- Master in Medical Microbiology, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Jorge Arturo Parra Montoya
- Servicio de Ginecología y Obstetricia, Hospital Civil Juan I. Menchaca, Guadalajara 44340, Mexico; (J.A.P.M.); (G.S.R.)
| | - Geraldine Sosa Romero
- Servicio de Ginecología y Obstetricia, Hospital Civil Juan I. Menchaca, Guadalajara 44340, Mexico; (J.A.P.M.); (G.S.R.)
| | - Lizbeth de Jesús González Piñuelas
- Laboratorio de Diagnóstico de Enfermedades Emergentes y Reemergentes, Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (L.d.J.G.P.); (N.V.M.)
| | - Adriana M. López-Barradas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (A.M.L.-B.); (O.G.-P.)
| | - Omar Granados-Portillo
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico; (A.M.L.-B.); (O.G.-P.)
| | - Mariel García Chagollán
- Instituto de Investigación de Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.G.C.); (A.L.P.S.)
| | - Ana Laura Pereira Suárez
- Instituto de Investigación de Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.G.C.); (A.L.P.S.)
| | - Patrick M. Gillevet
- Microbiome Analysis Center, George Mason University, Manassas, VA 20110, USA;
| | - Natali Vega Magaña
- Laboratorio de Diagnóstico de Enfermedades Emergentes y Reemergentes, Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (L.d.J.G.P.); (N.V.M.)
- Instituto de Investigación de Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.G.C.); (A.L.P.S.)
| | - Marcela Peña Rodríguez
- Laboratorio de Diagnóstico de Enfermedades Emergentes y Reemergentes, Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (L.d.J.G.P.); (N.V.M.)
| |
Collapse
|
38
|
Vakili S, Koohpeyma F, Samare-Najaf M, Jahromi BN, Jafarinia M, Samareh A, Hashempur MH. The Effects of L-Tartaric Acid on Ovarian Histostereological and Serum Hormonal Analysis in an Animal Model of Polycystic Ovary Syndrome. Reprod Sci 2024; 31:3583-3594. [PMID: 39333435 DOI: 10.1007/s43032-024-01699-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine-related reproductive disorder in women of reproductive age, accompanied by both the impairment of female fecundity and a risk of metabolic disorders. PCOS is emphasized as a worldwide concern due to its unknown etiology and lack of specific medications. The current study aimed to evaluate the effects of L-tartaric acid, an abundantly occurring compound in fruits, on the histostereological and hormonal changes caused by PCOS. Forty adult Sprague Dawley rats were randomly divided into four groups including controls (no intervention), Tartaric acid (40mg/Kg/day from day 21 onwards for 39 days), PCOS (21 days letrozole and then normal saline orally for 39 days), and PCOS + Tartaric acid. After treatments, the ovarian histostereological analysis as well as the level of reproductive hormones including luteinizing hormone (LH), follicle-stimulating hormone (FSH), estradiol, progesterone, and testosterone was measured. PCOS caused a significant decrease in the number of unilaminar, multilaminar, antral, and graafian follicles and increased follicular atresia (p-value < 0.001). Moreover, the weight and volume of ovarian tissue and related structures including cortex, medulla, and cysts increased significantly (p-value < 0.0001). However, corpus luteum volume was significantly decreased (p-value < 0.001). Although significant differences were found in some parameters with the control group (p-value < 0.05), the administration of tartaric acid restored the pathological effects of PCOS on the ovarian histostructure. Furthermore, tartaric acid improved the serum levels of LH, estradiol, progesterone, and testosterone (p-value < 0.05). The obtained findings may suggest tartaric acid as a novel strategy for PCOS management, although further studies are necessary.
Collapse
Affiliation(s)
- Sina Vakili
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farhad Koohpeyma
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammad Samare-Najaf
- 3Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Bahia Namavar Jahromi
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of OB-GYN, Division of Infertility and IVF, Shiraz School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Morteza Jafarinia
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Samareh
- Department of Clinical Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Hashem Hashempur
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
39
|
Shangguan F, Liu H, Guo Y, Yu J, Liang Y, Yu H, Su Y, Li Z. Effectiveness and acceptability of different lifestyle interventions for women with polycystic ovary syndrome: protocol for a systematic review and network meta-analysis. BMJ Open 2024; 14:e084815. [PMID: 39477270 PMCID: PMC11529581 DOI: 10.1136/bmjopen-2024-084815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
INTRODUCTION Women with polycystic ovary syndrome (PCOS) experience various metabolic, endocrine, reproductive and psychosocial manifestations. Lifestyle modification is crucial for the management of PCOS to reduce long-term complications. Nonetheless, the efficacy and acceptability of lifestyle interventions differs, and there are no uniform methods of clinical application. Hence, a systematic review and network meta-analysis (NMA) are needed to explore the efficacy and acceptability of lifestyle interventions to inform clinical practice. METHODS AND ANALYSIS Ten databases (Cochrane Gynaecology and Fertility Specialised Register, Cochrane Register of Studies Online, PubMed, EMBASE (Excerpta Medica Database), PsycINFO, CINAHL (Cumulative Index to Nursing and Allied Health Literature), Chinese National Knowledge Infrastructure, WanFang, VIP, and Sinomed) and four clinical trial registry platforms will be searched to identify literature published in English or Chinese reporting results of randomised clinical trials conducted to evaluate the effects of lifestyle interventions for women with PCOS. The reference lists of the included studies will be manually searched. Primary outcomes will include biochemical and clinical hyperandrogenism, recruitment and retention rates. Secondary outcomes will encompass menstrual regularity, ovulation, anthropometry and quality of life. Literature selection and extraction of data will be performed independently by at least two researchers. An NMA random-effects model will be implemented for amalgamating evidence. All treatments will be ranked based on the value of p. OpenBUGS will be used for Bayesian modelling, with output verifications generated in Stata and R. The quality of evidence supporting network estimates of major outcomes will also be appraised using the Grading of Recommendations Assessment, Development, and Evaluation framework. ETHICS AND DISSEMINATION Ethical approval is not required for this review as no data will be collected from human participants. Results will be presented in a peer-reviewed publication. PROSPERO REGISTRATION NUMBER CRD42024499819.
Collapse
Affiliation(s)
| | - Hui Liu
- University of South China School of Nursing, Hengyang, Hunan, China
| | - Yu Guo
- University of South China School of Nursing, Hengyang, Hunan, China
| | - Juping Yu
- Faculty of Life Sciences and Education, University of South Wales, Pontypridd, UK
| | - Yinni Liang
- University of South China School of Nursing, Hengyang, Hunan, China
| | - Huixi Yu
- University of South China School of Nursing, Hengyang, Hunan, China
| | - Yinhua Su
- University of South China School of Nursing, Hengyang, Hunan, China
| | - Zhongyu Li
- University of South China School of Nursing, Hengyang, Hunan, China
| |
Collapse
|
40
|
Hellberg A, Salamon D, Ujvari D, Rydén M, Hirschberg AL. Weight Changes Are Linked to Adipose Tissue Genes in Overweight Women with Polycystic Ovary Syndrome. Int J Mol Sci 2024; 25:11566. [PMID: 39519120 PMCID: PMC11547111 DOI: 10.3390/ijms252111566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Women with polycystic ovary syndrome (PCOS) have varying difficulties in achieving weight loss by lifestyle intervention, which may depend on adipose tissue metabolism. The objective was to study baseline subcutaneous adipose tissue gene expression as a prediction of weight loss by lifestyle intervention in obese/overweight women with PCOS. This is a secondary analysis of a randomized controlled trial where women with PCOS, aged 18-40 and body mass index (BMI) ≥ 27 were initially randomized to either a 4-month behavioral modification program or minimal intervention according to standard care. Baseline subcutaneous adipose tissue gene expression was related to weight change after the lifestyle intervention. A total of 55 obese/overweight women provided subcutaneous adipose samples at study entry. Weight loss was significant after behavioral modification (-2.2%, p = 0.0014), while there was no significant weight loss in the control group (-1.1%, p = 0.12). In microarray analysis of adipose samples, expression of 40 genes differed significantly between subgroups of those with the greatest weight loss or weight gain. 10 genes were involved in metabolic pathways including glutathione metabolism, gluconeogenesis, and pyruvate metabolism. Results were confirmed by real-time polymerase chain reaction (RT-PCR) in all 55 subjects. Expressions of GSTM5, ANLN, and H3C2 correlated with weight change (R = -0.41, p = 0.002; R = -0.31, p = 0.023 and R = -0.32, p = 0.016, respectively). GSTM5, involved in glutathione metabolism, was the strongest predictor of weight loss, and together with baseline waist-hip ratio (WHR) explained 31% of the variation in body weight change. This study shows that baseline subcutaneous adipose tissue genes play a role for body weight outcome in response to lifestyle intervention in overweight/obese women with PCOS.
Collapse
Affiliation(s)
- Anton Hellberg
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
| | - Daniel Salamon
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
| | - Dorina Ujvari
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
- Department of Microbiology, Tumor and Cell Biology, National Pandemic Centre, Centre for Translational Microbiome Research, Karolinska Institutet, 17165 Solna, Sweden
| | - Mikael Rydén
- Department of Medicine Huddinge (H7), Karolinska Institutet, C2-94 Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden;
| | - Angelica Lindén Hirschberg
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital Solna, 17176 Stockholm, Sweden
| |
Collapse
|
41
|
Xue H, Hu Z, Liu S, Zhang S, Yang W, Li J, Yan C, Zhang J, Zhang J, Lei X. The mechanism of NF-κB-TERT feedback regulation of granulosa cell apoptosis in PCOS rats. PLoS One 2024; 19:e0312115. [PMID: 39453929 PMCID: PMC11508119 DOI: 10.1371/journal.pone.0312115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/13/2024] [Indexed: 10/27/2024] Open
Abstract
Patients with Polycystic ovary syndrome (PCOS) have chronic low-grade ovarian inflammation. Inflammation can cause telomere dysfunction, and telomere and telomerase complex are also involved in regulating inflammation. However, the specific mechanisms of inflammatory signaling feedback and telomere-telomerase mutual regulation remain to be discovered. This study elucidates the role of Nuclear factor kappa-B (NF-κB)-Telomerase reverse transcriptase (TERT) feedback in PCOS granulosa cell apoptosis. Using letrozole and a high-fat diet, a PCOS rat model was established, along with a Lipopolysaccharide (LPS) -treated KGN cell inflammation model was established. NF-κB and TERT inhibitors (BAY 11-7082 and BIBR1532) were then administered to LPS-induced KGN cells. PCOS rats displayed disrupted estrous cycles, increased weight, elevated serum testosterone, cystic follicles, granulosa cell layer thinning, and reduced corpora lutea count (P are all less than 0.05). In PCOS rat ovaries, NF-κB, Interleukin-6 (IL-6), Tumor Necrosis Factor α (TNF-α), TERT, Bax, and Caspase-3 exhibited notable upregulation, while Bcl-2 decreased, with telomere elongation (P are all less than 0.05). There were significant correlations among NF-κB-related inflammatory factors, TERT and apoptotic factors, and they were positively correlated with Bax and Caspase-3, and negatively correlated with Bcl-2 (P are all less than 0.05). LPS-treated KGN cells demonstrated increased expression of inflammatory and pro-apoptotic factors, later restored post-treatment with NF-κB and TERT inhibitors (P are all less than 0.05). In conclusion, TERT may induce granulosa cell apoptosis by participating in the regulation of the NF-κB signaling pathway, thereby mediating the chronic inflammatory response of PCOS through downstream inflammatory factors IL-6 and TNF-α.
Collapse
Affiliation(s)
- Haoxuan Xue
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Clinical Anatomy and Reproductive Medicine Application Institute, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zecheng Hu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shun Liu
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Clinical Anatomy and Reproductive Medicine Application Institute, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shun Zhang
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Wenqin Yang
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Clinical Anatomy and Reproductive Medicine Application Institute, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiasi Li
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Clinical Anatomy and Reproductive Medicine Application Institute, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chulin Yan
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Clinical Anatomy and Reproductive Medicine Application Institute, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiaming Zhang
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Clinical Anatomy and Reproductive Medicine Application Institute, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jing Zhang
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Clinical Anatomy and Reproductive Medicine Application Institute, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaocan Lei
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Clinical Anatomy and Reproductive Medicine Application Institute, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
42
|
Cheng T, Shi H, Bu Z, Yu Y, Song W, Haixia J, Yao G. Contribution of cervical incompetence to occurrence of second trimester abortion in patients with polycystic ovary syndrome during the frozen embryo transfer cycle. Front Endocrinol (Lausanne) 2024; 15:1411618. [PMID: 39479270 PMCID: PMC11521832 DOI: 10.3389/fendo.2024.1411618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Background Second-trimester abortion is a critical issue in infertile women with polycystic ovary syndrome (PCOS) treated with assisted reproductive technology (ART), cervical incompetence (CI) may play a role. Although previous studies have revealed an association between PCOS and CI in women undergoing ART with fresh embryo transfer, the associated risk factors in frozen embryo transfer cycles are still unknown. The primary objective of this study is to examine the impact of CI on the occurrence of second-trimester abortion in women with PCOS undergoing frozen embryo transfer. Methods This retrospective cohort study included patients who underwent frozen-thawed embryo transfer and experienced second-trimester abortion between January 2012 and January 2020 from the Reproductive Medical Center of the First Affiliated Hospital of Zhengzhou University. Logit-transformed propensity score matching (PSM) was used to assess covariates. Patients were classified into the PCOS and non-PCOS groups. The PCOS group was further divided into two subgroups: the CI group and non-CI group. Results After case matching with PSM, 278 patients were included: 139 each in the PCOS group and non-PCOS groups. More miscarriages were attributed to CI in the PCOS group compared with the control group (20.14% vs. 10.07%). Subsequently, in the PCOS group, CI and non-CI subgroup analyses revealed a higher transfer cleavage-stage embryo incidence in the CI group than in the blastocysts group (p=0.001). Moreover, the between-group miscarriage-related gestational age varied significantly (p=0.039). Binary logistic regression analysis revealed that cleavage embryo transfer (p= 0.047) was associated with increased CI risk in the PCOS group, besides, increasing the number of transferred embryos did not impact the occurrence of CI in patients with PCOS. Conclusion CI independently predicted a higher risk of second-trimester abortion in patients with PCOS during the frozen embryo transfer cycle. What's more, increasing the number of transferred embryos did not affect the incidence of CI in the PCOS group.
Collapse
Affiliation(s)
- Tingting Cheng
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hao Shi
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiqin Bu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiping Yu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenyan Song
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin Haixia
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guidong Yao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
43
|
Liao B, Chen W, Qi X, Yun C, Pang Y. Interleukin-22 improves ovulation in polycystic ovary syndrome via STAT3 signaling. Mol Hum Reprod 2024; 30:gaae037. [PMID: 39423135 DOI: 10.1093/molehr/gaae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 09/23/2024] [Indexed: 10/21/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disease, which leads to serious impairment of reproductive health in women of child-bearing age. Anovulation or oligo-ovulation is a common clinical manifestation of PCOS patients. A disturbance of the ovarian immune microenvironment contributes to the disorders of follicle development and ovulation; however, the underlying mechanism remains unclear. Here we demonstrated the protective effect of immune factor interleukin-22 (IL-22) on PCOS follicle development and ovulation. Follicular IL-22 levels were significantly lower in PCOS patients than in the control group and were positively correlated with oocyte fertilization rate and high-quality embryo rate. Additionally, IL-22 evidently improved follicle development in vitro and promoted ovulation-related gene expression, which was disrupted by the depletion of interleukin-22 receptor 1 (IL-22R1) or inhibition of STAT3 in granulosa cells. This indicates that IL-22 acts through IL-22R1 and the STAT3 signaling pathway to promote follicle development and ovulation in PCOS. In summary, this study has elucidated the vital role of the ovarian immune microenvironment in follicle development and ovulation. Application of IL-22 may provide new insights into the treatment of PCOS patients.
Collapse
Affiliation(s)
- Baoying Liao
- Department of Obstetrics and Gynaecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
| | - Weixuan Chen
- Department of Obstetrics and Gynaecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
| | - Xinyu Qi
- Department of Obstetrics and Gynaecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Chuyu Yun
- Department of Obstetrics and Gynaecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yanli Pang
- Department of Obstetrics and Gynaecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| |
Collapse
|
44
|
Orosz M, Borics F, Rátonyi D, Vida B, Csehely S, Jakab A, Lukács L, Lampé R, Deli T. Pre-Conception Androgen Levels and Obstetric Outcomes in Polycystic Ovary Syndrome: A Single-Center Retrospective Study. Diagnostics (Basel) 2024; 14:2241. [PMID: 39410647 PMCID: PMC11476020 DOI: 10.3390/diagnostics14192241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Hyperandrogenism is a determining diagnostic factor for PCOS. If pregnancy is conceived, it is considered high-risk due to several potential complications, but the correlation between pre-pregnancy androgen levels and obstetric outcomes is poorly characterized. OBJECTIVE To determine if pre-pregnancy serum androgen concentrations and androgen indexes differed when certain obstetric and neonatal outcomes appeared in PCOS. METHODS A single-center, retrospective study was carried out. All patients were treated between 2012 and 2019. A total of 73 patients had all the endocrine and obstetric data available. Pre-pregnancy hormone levels (total testosterone-T, androstenedione-AD, DHEAS (dehydroepiandrosterone sulfate), SHBG (sex-hormone-binding globulin), and TSH (thyroid-stimulating hormone) were collected, and T/SHBG, AD/SHBG, DHEAS/SHBG, T/AD indexes were calculated and compared. RESULTS When miscarriage was present in the history, significantly elevated pre-pregnancy AD levels were observed. Higher pre-pregnancy AD level was noted in PCOS patients delivering female newborns as compared to males. Additionally, a higher T/AD ratio was associated with subsequent preterm delivery, but significance was lost after age adjustment. Maternal age at delivery had a significant negative correlation with pre-pregnancy DHEAS levels and DHEAS/SHBG ratio. Pre-pregnancy SHBG displayed a significant negative correlation, while pre-pregnancy androgen/SHBG ratios exhibited positive correlations with both birth weight and birth weight percentile. CONCLUSIONS Based on our data, AD and the T/AD ratio emerge as distinctive factors in certain outcomes, implying a potential specific role of altered 17-β-HSD (17β-hydroxysteroid dehydrogenase) enzyme activity, possibly influencing offspring outcomes. The pre-pregnancy T/SHBG ratio exhibits a potentially stronger correlation with fetal growth potential compared to SHBG alone. DHEAS and maternal age at delivery are strongly correlated in PCOS patients.
Collapse
Affiliation(s)
- Mónika Orosz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary; (M.O.); (S.C.)
| | - Fanni Borics
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary;
| | - Dávid Rátonyi
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary; (M.O.); (S.C.)
| | - Beáta Vida
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary; (M.O.); (S.C.)
| | - Szilvia Csehely
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary; (M.O.); (S.C.)
| | - Attila Jakab
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary; (M.O.); (S.C.)
| | - Luca Lukács
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary; (M.O.); (S.C.)
| | - Rudolf Lampé
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary; (M.O.); (S.C.)
| | - Tamás Deli
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary; (M.O.); (S.C.)
| |
Collapse
|
45
|
Teede HJ, Mousa A, Tay CT, Costello MF, Brennan L, Norman RJ, Pena AS, Boyle JA, Joham A, Berry L, Moran L. Summary of the 2023 international evidence-based guideline for the assessment and management of polycystic ovary syndrome: an Australian perspective. Med J Aust 2024; 221:389-395. [PMID: 39223729 DOI: 10.5694/mja2.52432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 06/17/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION The Australian-led 2023 International evidence-based guideline for the assessment and management of polycystic ovary syndrome was based on best available evidence, clinical expertise and consumer preference. It followed best practice, involved extensive evidence synthesis and applied relevant frameworks across evidence quality, feasibility, acceptability, cost and implementation. Thirty-nine societies and organisations covering 71 countries were engaged. The evidence in the assessment and management of polycystic ovary syndrome (PCOS) has generally improved in the past five years, but remains of low to moderate quality. The technical evidence report, 52 systematic reviews and analyses (approximately 6000 pages) underpin 77 evidence-based and 54 consensus recommendations, with 123 practice points. MAIN RECOMMENDATIONS Changes include: refinement of individual diagnostic criteria, a simplified diagnostic algorithm and inclusion of anti-Müllerian hormone levels as an alternative to ultrasound in adults only, and differentiation of adolescent and adult criteria; strengthening the recognition of broad features of PCOS including metabolic effects, cardiovascular disease, dermatological symptoms, sleep apnoea, a high prevalence of psychological features and a high risk of adverse pregnancy outcomes; emphasising the poorly recognised, diverse burden of disease, the vital need for greater health professional education, evidence-based patient information, improved models of care, shared decision making and research efforts to improve patient experience; maintained emphasis on healthy lifestyle, emotional wellbeing and quality of life, with awareness and consideration of weight stigma; and emphasising evidence-based medical therapy and cheaper and safer fertility management. CHANGES IN MANAGEMENT AS A RESULT OF THIS GUIDELINE The 2023 guideline is approved by the National Health and Medical Research Council and provides clinicians and patients with clear advice on best practice in a common and neglected condition, based on the best available evidence, expert multidisciplinary input and consumer preferences. It provides vital, extensive patient and provider resources to enhance evidence-based care. The full guideline is available at www.monash.edu/medicine/mchri/pcos/guideline.
Collapse
Affiliation(s)
- Helena J Teede
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, VIC
| | - Aya Mousa
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, VIC
| | - Chau T Tay
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, VIC
| | - Michael F Costello
- University of New South Wales, Sydney, NSW
- Royal Hospital for Women, Sydney, NSW
- Monash IVF, Sydney, NSW
| | | | - Robert J Norman
- Robinson Research Institute, University of Adelaide, Adelaide, SA
| | - Alexia S Pena
- Robinson Research Institute, University of Adelaide, Adelaide, SA
| | | | - Anju Joham
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, VIC
| | - Lorna Berry
- Polycystic Ovary Syndrome Association of Australia, Sydney, NSW
| | - Lisa Moran
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, VIC
- Robinson Research Institute, University of Adelaide, Adelaide, SA
| |
Collapse
|
46
|
Wydra J, Szlendak-Sauer K, Zgliczyńska M, Żeber-Lubecka N, Ciebiera M. Gut Microbiota and Oral Contraceptive Use in Women with Polycystic Ovary Syndrome: A Systematic Review. Nutrients 2024; 16:3382. [PMID: 39408349 PMCID: PMC11478613 DOI: 10.3390/nu16193382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Polycystic ovary syndrome (PCOS) is one of the most prevalent endocrine syndromes affecting women at reproductive age. With increasing knowledge of the role of the microbiota in the pathogenesis of PCOS, new management strategies began to emerge. However, data on the impact of established treatment regimens, such as metformin and oral contraceptive agents, on the gut microbiota composition are scarce. This study aimed to evaluate the specificity of the gut microbiota in women with PCOS before and after treatment with oral contraceptives. Methods: We have systematically searched the following databases: PubMed/MEDLINE, Scopus, Web of Science and Google Scholar. The last search was performed on 13 May 2024. We included only full-text original research articles written in English. The risk of bias was assessed using a modified version of the Newcastle-Ottawa Scale. Results: The above described search strategy retrieved 46 articles. Additionally, 136 articles were identified and screened through Google Scholar. After removing duplicates, we screened the titles and abstracts, resulting in three eligible articles constituting the final pool. They were published between 2020 and 2022 and are based on three ethnically distinct study populations: Turkish, Spanish and American. The studies included a total of 37 women diagnosed with PCOS and using OCs. Conclusions: OC treatment does not seem to affect the gut microbiota in a significant way in patients with PCOS in short observation. Well-designed randomized controlled studies with adequate, unified sample size are lacking.
Collapse
Affiliation(s)
- Jakub Wydra
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-809 Warsaw, Poland
| | - Katarzyna Szlendak-Sauer
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 00-189 Warsaw, Poland
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland
| | - Magdalena Zgliczyńska
- Department of Obstetrics, Perinatology and Neonatology, Centre of Postgraduate Medical Education, 01-809 Warsaw, Poland
| | - Natalia Żeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
| | - Michał Ciebiera
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 00-189 Warsaw, Poland
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland
| |
Collapse
|
47
|
Miao J, Gao L, Liu X, Cai W, Chen L, Chen M, Sun Y. Exploring the therapeutic mechanisms of Yikang decoction in polycystic ovary syndrome: an integration of GEO datasets, network pharmacology, and molecular dynamics simulations. Front Med (Lausanne) 2024; 11:1455964. [PMID: 39421869 PMCID: PMC11484630 DOI: 10.3389/fmed.2024.1455964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Objective The incidence of Polycystic Ovary Syndrome (PCOS) is increasing annually. This study aims to investigate the therapeutic mechanisms of Yikang Decoction (YKD) in the treatment of PCOS through the integration of GEO datasets, network pharmacology, and dynamic simulation. Methods Active ingredients of YKD and their targets were collected from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) platform. Disease-relevant targets for PCOS were retrieved from several databases, including GeneCards, OMIM, PharmGKB, DrugBank, and GEO. The underlying pathways associated with the overlapping targets between YKD and PCOS were identified using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. The mechanisms of interaction between the core targets and components were further explored through molecular docking and molecular dynamics simulations (MD). Results 139 potential active components and 315 targets of YKD were identified. A topological analysis of the PPI network revealed 10 core targets. These targets primarily participated in the regulation of biological processes, including cell metabolism, apoptosis, and cell proliferation. The pathways associated with treating PCOS encompassed PI3K-Akt signaling pathway, Lipid and atherosclerosis, MAPK signaling pathways, and Endocrine resistance signaling pathways. Moreover, molecular docking and MD have been shown to reveal a good binding capacity between active compounds and screening targets. Conclusion This study systematically investigates the multi-target mechanisms of YKD in the treatment of PCOS, with preliminary verification provided through molecular docking and MD. The findings offer compelling evidence supporting the efficacy of YKD in treating PCOS.
Collapse
Affiliation(s)
- Jiang Miao
- Department of Pharmacy, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| | - LiXuan Gao
- Department of Rehabilitation Medicine, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| | - Xi Liu
- Wuyanling National Natural Reserve Administrative of Zhejiang, Wenzhou, China
| | - Wenpin Cai
- Department of Laboratory Medicine, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| | - Lei Chen
- Department of Pharmacy, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| | - Mojinzi Chen
- Department of Chinese Internal Medicine, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| | - Yun Sun
- Department of Gynaecology, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| |
Collapse
|
48
|
Gao X, Wang Y, Wang Y, Yang Z, Yan X, Li S, Jiang Y, Li Y, Zhao S, Zhao H, Chen ZJ. No Association of Polycystic Ovary Syndrome with Pancreatic Cancer: A Mendelian Randomization Study. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:522-524. [PMID: 39723230 PMCID: PMC11666843 DOI: 10.1007/s43657-024-00156-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 12/28/2024]
Abstract
Recently, there has been a debate regarding the association between polycystic ovary syndrome (PCOS) and pancreatic cancer (PC). In order to examine the causal relationship between PCOS and PC, we conducted a Mendelian randomization study, which utilized 12 single nucleotide polymorphisms (SNPs) identified from a genome-wide association study (GWAS) meta-analysis that included 10,074 PCOS cases and 103,164 controls of European ancestry as instrumental variables (IVs). The outcome data were obtained from the FinnGen database (including 605 cases and 218,187 controls). We demonstrate that genetically predicted PCOS is not causally associated with PC risk in Europeans (odds ratio = 0.99, 95% confidence interval (CI) = 0.72-1.36, p > 0.05). Sensitivity analysis showed horizontal pleiotropy (intercept p > 0.05), heterogeneity (Cochran Q p > 0.05), and the leave-one-out sensitivity test showed that individual SNP effects had no influence on the results. In conclusion, our study did not provide evidence of a causal link between PCOS and PC. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-024-00156-y.
Collapse
Affiliation(s)
- Xueying Gao
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127 China
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127 China
| | - Yuteng Wang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
| | - Yikun Wang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
| | - Ziyi Yang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
| | - Xueqi Yan
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
| | - Shumin Li
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127 China
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127 China
| | - Yonghui Jiang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
| | - Yimeng Li
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
| | - Shigang Zhao
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
| | - Han Zhao
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
| | - Zi-Jiang Chen
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012 Shandong China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012 Shandong China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, 250012 Shandong China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127 China
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127 China
| |
Collapse
|
49
|
Li Y, Zhang J, Zheng X, Lu W, Guo J, Chen F, Liu C. Depression, anxiety and self-esteem in adolescent girls with polycystic ovary syndrome: a systematic review and meta-analysis. Front Endocrinol (Lausanne) 2024; 15:1399580. [PMID: 39403587 PMCID: PMC11471625 DOI: 10.3389/fendo.2024.1399580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/10/2024] [Indexed: 11/02/2024] Open
Abstract
Objective Studies have shown the adverse psychological impact of polycystic ovary syndrome (PCOS), but the state of mental health in adolescents with PCOS remains unclear. Thus, we performed a systematic review and meta-analysis to investigate the prevalence and severity of depression and anxiety, as well as potential effects on self-esteem and quality of life (QoL) in this specific population. Methods We systematically searched four electronic databases: PubMed, Embase, Web of Science, and the Cochrane Reviews database for articles published until 25/8/2024. We considered observational studies in which the subjects were adolescent girls with PCOS who had reported symptoms including anxiety, depression, self-esteem, and QoL. The Review Manager version 5.4 was used to analyze the available data extracted. We used the Newcastle-Ottawa Quality Assessment Scale (NOS) to evaluate the quality of selected studies. A funnel plot was utilized to assess the risk of literature bias, and a forest plot was used to represent the combined outcomes. This systematic review was previously registered in PROSPERO with the registration number CRD42022382036. Results We included 11 studies in the systematic review and conducted meta-analyses on 10 of them. Adolescents with PCOS reported a higher risk of depression (OR = 2.21, 95% CI: 1.23 to 4.00, p = 0.008) and a higher level of depression scores (SMD = 0.43, 95% CI: 0.16 to 0.71, p = 0.002) than controls. There were no significant differences in anxiety (OR = 1.90, 95% CI: 0.52 to 6.96, p = 0.33; SMD = 0.19, 95% CI: -0.21 to 0.59, p = 0.36), self-esteem (SMD = -0.17, 95% CI: -0.85 to 0.52, p = 0.64), and QoL (SMD = -0.15, 95% CI: -0.42 to 0.11, p = 0.26) between the two groups. Conclusions Our research indicated that adolescents with PCOS experienced more severe depressive symptoms than those without PCOS. This highlights the importance of evaluation and early treatment of mental health in PCOS. More clinicians should pay attention to the mental health of adolescent girls with PCOS through this study. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022382036.
Collapse
Affiliation(s)
- Yuxin Li
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiayu Zhang
- Department of Nursing, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xuanling Zheng
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenjing Lu
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jinru Guo
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Fuhong Chen
- Department of Nursing, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Changqin Liu
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Clinical Efficacy and Evidence Studies of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
50
|
Santander N, Figueroa EG, González-Candia A, Maliqueo M, Echiburú B, Crisosto N, Salas-Pérez F. Oxidative Stress in Polycystic Ovary Syndrome: Impact of Combined Oral Contraceptives. Antioxidants (Basel) 2024; 13:1168. [PMID: 39456422 PMCID: PMC11505064 DOI: 10.3390/antiox13101168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Polycystic Ovary Syndrome (PCOS) is a complex hormonal disorder that is associated with heightened metabolic risks. While oxidative stress (OS) is known to play a role in PCOS, the precise nature of the relationship between PCOS and increased OS remains not entirely understood. Combined oral contraceptives (COCs) are the first-line treatment to regulate menstrual cycles and androgen levels, but their impact on oxidative stress requires further study. We conducted a transcriptomic analysis using RNAseq and assessed the levels of various oxidative stress (OS) markers in serum samples from women with PCOS and controls and whether they were using combined oral contraceptives (COCs), including enzymatic activities, FRAP, and 8-isoprostane (8-iso). A total of 359 genes were differentially expressed in women with PCOS compared to control women. Genes differentially expressed were enriched in functions related to inflammation and, interestingly, oxidative stress response. In controls, 8-iso levels were increased in women using COCs, whereas in women with PCOS, 8-iso levels were reduced in those using oral contraceptives (191.1 ± 97 vs. 26.4 ± 21 pg/mL, p: <0.0001). Correlation analyses showed a trend for a negative correlation between 8-iso and Ferriman score in women with PCOS consuming COCs (r = -0.86, p = 0.06) and a negative correlation between GSH and hyperandrogenism in women with PCOS (r = -0.89, p = 0.01). These results reveal the presence of lipid peroxidation in women with PCOS, which was modified by the use of COCs, providing new insights into the pathophysiology of PCOS in the Chilean population.
Collapse
Affiliation(s)
- Nicolás Santander
- Health Sciences Institute, Universidad de O’Higgins, Rancagua 282000, Chile; (N.S.); (E.G.F.); (A.G.-C.)
| | - Esteban G. Figueroa
- Health Sciences Institute, Universidad de O’Higgins, Rancagua 282000, Chile; (N.S.); (E.G.F.); (A.G.-C.)
| | - Alejandro González-Candia
- Health Sciences Institute, Universidad de O’Higgins, Rancagua 282000, Chile; (N.S.); (E.G.F.); (A.G.-C.)
| | - Manuel Maliqueo
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine, West Division, Faculty of Medicine, Universidad de Chile, Santiago 8350499, Chile; (M.M.); (B.E.); (N.C.)
| | - Bárbara Echiburú
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine, West Division, Faculty of Medicine, Universidad de Chile, Santiago 8350499, Chile; (M.M.); (B.E.); (N.C.)
| | - Nicolás Crisosto
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine, West Division, Faculty of Medicine, Universidad de Chile, Santiago 8350499, Chile; (M.M.); (B.E.); (N.C.)
- Endocrinology Endocrinology Unit, Department of Medicine, Clínica Alemana de Santiago, Faculty of Medicine, Universidad del Desarrollo, Santiago 7650568, Chile
| | - Francisca Salas-Pérez
- Health Sciences Institute, Universidad de O’Higgins, Rancagua 282000, Chile; (N.S.); (E.G.F.); (A.G.-C.)
| |
Collapse
|