201
|
Kim CH, Lee SH. Effectiveness of Lifestyle Modification in Polycystic Ovary Syndrome Patients with Obesity: A Systematic Review and Meta-Analysis. Life (Basel) 2022; 12:life12020308. [PMID: 35207595 PMCID: PMC8876590 DOI: 10.3390/life12020308] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 12/20/2022] Open
Abstract
(1) Background: Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility and endocrine disorders among women of reproductive age. Previous studies have employed lifestyle interventions to manage anovulatory infertility and endocrine disorders. However, the effect of lifestyle interventions on the metabolic index remains ambiguous; (2) Methods: Data were obtained through a systematic search of the Ovid-Medline, Ovid-EMBASE, and Cochrane Library databases. Two reviewers independently reviewed the literature in two stages. A consensus was achieved through discussions regarding the final selection of the literature; (3) Results: This study observed that the group that underwent lifestyle modifications displayed significant improvement in reproductive function compared to the control group. Combination therapy with diet and exercise resulted in improved fasting insulin levels, compared to monotherapy with diet or exercise. Moreover, moderate weight loss (a minimum of 5%) resulted in an improved metabolic index. The subgroup analysis revealed that the group that underwent lifestyle modifications had a significantly higher number of patients with improved menstrual cycles, compared to the control groups; (4) Conclusions: Lifestyle modification using combination therapy is a promising therapeutic approach that can be employed in the management of PCOS patients with obesity. This scenario warrants further studies with larger sample sizes to develop ideal treatment protocols.
Collapse
Affiliation(s)
- Chan-Hee Kim
- Artificial Kidney Team, Incheon Sejong Hospital, Incheon 21080, Korea;
| | - Seon-Heui Lee
- Department of Nursing Science, College of Nursing, Gachon University, Incheon 21936, Korea
- Correspondence: ; Tel.: +82-10-9369-7326
| |
Collapse
|
202
|
Ilagan-Vega MKC, Tantengco OAG, Paz-Pacheco E. A bibliometric analysis of polycystic ovary syndrome research in Southeast Asia: Insights and implications. Diabetes Metab Syndr 2022; 16:102419. [PMID: 35152063 DOI: 10.1016/j.dsx.2022.102419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND AIMS There has been an increased understanding in the pathophysiology and management of polycystic ovary syndrome (PCOS) in recent years. This study aimed to evaluate the trends and characteristics of PCOS publications in Southeast Asia (SEA) through a bibliometric analysis. METHODS Systematic review of literature on PCOS in SEA countries between 1975 and 2020 was performed using the Scopus database. All published studies on PCOS conducted in or published by authors from any of the countries in SEA were included in this analysis. Bibliographic information was obtained, and visualization of collaboration networks of countries and keywords was conducted using VOSviewer software. RESULTS A total of 260 articles were included in this analysis. The number of PCOS publications in SEA continued to increase through the years. The country with the highest number of publications was Thailand while Singapore produced the most publications with the greatest impact. The focus of research in SEA was on PCOS prevalence, risk factors, infertility, and metabolic complications. A country's fertility rate, research and development expenditures (%GDP), number of researchers per million people, and international research collaborations significantly correlated with PCOS research impact in SEA. CONCLUSION This is the first bibliometric analysis in PCOS research in SEA. Despite the continuing increase in PCOS publications in SEA, it still lags compared to other regions. Countries in SEA should consider policies and strategies directed at increasing research support and encouraging international collaborations for local researchers to produce more meaningful publications and augment PCOS knowledge in the region.
Collapse
Affiliation(s)
| | | | - Elizabeth Paz-Pacheco
- Section of Endocrinology, Diabetes, and Metabolism, The Medical City, Ortigas Avenue, Pasig, Philippines; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of the Philippines - Philippine General Hospital, Taft Avenue, Manila, Philippines
| |
Collapse
|
203
|
Hu M, Zhang Y, Lu L, Zhou Y, Wu D, Brännström M, Shao LR, Billig H. Overactivation of the androgen receptor exacerbates gravid uterine ferroptosis via interaction with and suppression of the NRF2 defense signaling pathway. FEBS Lett 2022; 596:806-825. [DOI: 10.1002/1873-3468.14289] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/02/2021] [Accepted: 01/05/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Min Hu
- Department of Traditional Chinese Medicine The First Affiliated Hospital of Guangzhou Medical University 510120 Guangzhou China
- Institute of Integrated Traditional Chinese Medicine and Western Medicine Guangzhou Medical University 510120 Guangzhou China
- Department of Physiology/Endocrinology Institute of Neuroscience and Physiology The Sahlgrenska Academy University of Gothenburg 40530 Gothenburg Sweden
| | - Yuehui Zhang
- Department of Physiology/Endocrinology Institute of Neuroscience and Physiology The Sahlgrenska Academy University of Gothenburg 40530 Gothenburg Sweden
- Department of Obstetrics and Gynecology Key Laboratory and Unit of Infertility in Chinese Medicine First Affiliated Hospital Heilongjiang University of Chinese Medicine 150040 Harbin China
| | - Lingjing Lu
- Department of Traditional Chinese Medicine The First Affiliated Hospital of Guangzhou Medical University 510120 Guangzhou China
- Institute of Integrated Traditional Chinese Medicine and Western Medicine Guangzhou Medical University 510120 Guangzhou China
| | - Yu Zhou
- Department of Traditional Chinese Medicine The First Affiliated Hospital of Guangzhou Medical University 510120 Guangzhou China
- Institute of Integrated Traditional Chinese Medicine and Western Medicine Guangzhou Medical University 510120 Guangzhou China
| | - Denghui Wu
- Department of Obstetrics and Gynecology Key Laboratory and Unit of Infertility in Chinese Medicine First Affiliated Hospital Heilongjiang University of Chinese Medicine 150040 Harbin China
| | - Mats Brännström
- Department of Obstetrics and Gynecology Sahlgrenska University Hospital Sahlgrenska Academy University of Gothenburg 41345 Gothenburg Sweden
| | - Linus R Shao
- Department of Physiology/Endocrinology Institute of Neuroscience and Physiology The Sahlgrenska Academy University of Gothenburg 40530 Gothenburg Sweden
| | - Håkan Billig
- Department of Physiology/Endocrinology Institute of Neuroscience and Physiology The Sahlgrenska Academy University of Gothenburg 40530 Gothenburg Sweden
| |
Collapse
|
204
|
Spremović Rađenović S, Pupovac M, Andjić M, Bila J, Srećković S, Gudović A, Dragaš B, Radunović N. Prevalence, Risk Factors, and Pathophysiology of Nonalcoholic Fatty Liver Disease (NAFLD) in Women with Polycystic Ovary Syndrome (PCOS). Biomedicines 2022; 10:131. [PMID: 35052811 PMCID: PMC8773533 DOI: 10.3390/biomedicines10010131] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/26/2021] [Accepted: 01/03/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Polycystic Ovary Syndrome (PCOS) is one of the most common endocrine disorders in women's reproductive period of life. The presence of nonalcoholic fatty liver disease NAFLD, one of the leading causes of chronic liver disease in the Western world, is increased in women with PCOS. This review aims to present current knowledge in epidemiology, pathophysiology, diagnostics, and treatment of NAFLD in PCOS with an emphasis on the molecular basis of development of NAFLD in PCOS women. Methods: Authors investigated the available data on PCOS and NAFLD by a MEDLINE and Pub Med search during the years 1990-2021 using a combination of keywords such as "PCOS", "NAFLD", "steatohepatitis", "insulin resistance", "hyperandrogenaemia", "inflammation", "adipose tissue", and "obesity". Peer-reviewed articles regarding NAFLD and PCOS were included in this manuscript. Additional articles were identified from the references of relevant papers. Results: PCOS and NAFLD are multifactorial diseases, The development of NAFLD in PCOS women is linked to insulin resistance, hyperandrogenemia, obesity, adipose tissue dysfunction, and inflammation. There is the possible role of the gut microbiome, mitochondrial dysfunction, and endocannabinoid system in the maintenance of NAFLD in PCOS women. Conclusions: There is a need for further investigation about the mechanism of the development of NAFLD in PCOS women. New data about the molecular basis of development of NAFLD in PCOS integrated with epidemiological and clinical information could influence the evolution of new diagnostic and therapeutic approaches of NAFLD in PCOS.
Collapse
Affiliation(s)
- Svetlana Spremović Rađenović
- Clinic for Gynecology and Obstetrics, University Clinical Centre of Serbia, 11000 Belgrade, Serbia; (S.S.R.); (M.A.); (J.B.); (A.G.)
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Miljan Pupovac
- Clinic for Gynecology and Obstetrics, University Clinical Centre of Serbia, 11000 Belgrade, Serbia; (S.S.R.); (M.A.); (J.B.); (A.G.)
| | - Mladen Andjić
- Clinic for Gynecology and Obstetrics, University Clinical Centre of Serbia, 11000 Belgrade, Serbia; (S.S.R.); (M.A.); (J.B.); (A.G.)
| | - Jovan Bila
- Clinic for Gynecology and Obstetrics, University Clinical Centre of Serbia, 11000 Belgrade, Serbia; (S.S.R.); (M.A.); (J.B.); (A.G.)
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Svetlana Srećković
- Center for Anesthesiology and Resuscitation, University Clinical Centre of Serbia, 11000 Belgrade, Serbia;
| | - Aleksandra Gudović
- Clinic for Gynecology and Obstetrics, University Clinical Centre of Serbia, 11000 Belgrade, Serbia; (S.S.R.); (M.A.); (J.B.); (A.G.)
| | - Biljana Dragaš
- Intermedicus BIS, Specialized Hospital for Infertility, 11000 Belgrade, Serbia; (B.D.); (N.R.)
| | - Nebojša Radunović
- Intermedicus BIS, Specialized Hospital for Infertility, 11000 Belgrade, Serbia; (B.D.); (N.R.)
- Serbian Academy of Science and Art, 11000 Belgrade, Serbia
| |
Collapse
|
205
|
Zhang J, Ding N, Xin W, Yang X, Wang F. Quantitative Proteomics Reveals That a Prognostic Signature of the Endometrium of the Polycystic Ovary Syndrome Women Based on Ferroptosis Proteins. Front Endocrinol (Lausanne) 2022; 13:871945. [PMID: 35909514 PMCID: PMC9330063 DOI: 10.3389/fendo.2022.871945] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE We aimed to study the relationship between ferroptosis proteins and reproductive outcomes of infertile patients with PCOS and construct the related prognostic model. METHODS These endometrium samples of the study were collected from 33 women with PCOS and 7 control women with successful pregnancies at the Reproductive Center of Lanzhou University Second Hospital, September 2019 to September 2020. The 40 patients' endometrium was identified the differentially expressed proteins (DEPs) using liquid chromatography tandem mass spectrometry. The Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and Gene Ontology (GO) showed that the DEPs related pathways and functions between PCOS and controls. Subsequently, univariate Cox regression analysis and Lasso regression were used to identifying independent prognostic ferroptosis proteins, which were utilized to establish a prognostic model. Then the performance of the prognostic model was evaluated by receiver operating characteristic curve (ROC) and decision curve analysis (DCA). Then clinical data and prognostic model were used to predict the reproductive outcomes of PCOS patients by constructing the nomograms. Finally, we performed the single sample gene set enrichment analysis (ssGSEA) to explore the correlation between risk scores and immune status. RESULTS A total of 5331 proteins were identified, 391 proteins were differentially expressed in the PCOS and controls. The KEGG analysis revealed that the ferroptosis pathway was significantly different between PCOS and controls. 5 ferroptosis proteins (GPX4, DPP4, G6PD, PCBP1, and PCBP2) prognostic model (FerSig) was constructed via Cox regression and Lasso regression. Patients were separated into high and low-risk groups according to the FerSig. Kaplan-Meier curve showed that patients in the low-risk group had much better reproductive outcomes than those in the high-risk group. The DCA showed that the risk score was an independent predictive factor for reproductive outcomes. Compared with clinical data, ROC curve analysis indicated the FerSig proteins as a potential diagnostic and prognostic factor in PCOS patients. Functional analysis revealed that the FerSig proteins and immune microenvironment were correlated to the prognosis of PCOS. CONCLUSION The prognostic model focused on the FerSig proteins could predict the reproductive outcomes of PCOS patients with decreased endometrial receptivity, and provided theoretical basis for individualized treatment.
Collapse
|
206
|
Wang Y, Xiang T, Xia X, Zhang H, Geng S, Yang G, Qiu S, He Y, Liu R, Li L, Liu H, Li K, Zhang L, Liang Z, He J. Elevated circulating GPHB5 levels in women with insulin resistance and polycystic ovary syndrome: A cross-sectional study and multiple intervention studies. Front Endocrinol (Lausanne) 2022; 13:1010714. [PMID: 36568071 PMCID: PMC9772026 DOI: 10.3389/fendo.2022.1010714] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE GPHB5 has been found to be associated with glucose and lipid metabolism in animal studies. However, the association of GPHB5 with IR and metabolic disorders remains unknown, and there is a lack of research in humans. Our aim in this study was to investigate the relationship between circulating GPHB5 and metabolic disorders in humans. METHODS Bioinformatics analysis was performed to understand the relationship between GPHB5 and metabolic disorders. GPHB5 mRNA expression in mice and rats was determined using RT-qPCR. Circulating GPHB5 concentrations were measured with an ELISA kit. EHC and OGTT were performed in humans. RESULTS Bioinformatics analysis shows that GPHB5 is associated with metabolic disorders and PCOS. GPHB5 mRNA expression levels in the metabolic-related tissues of HFD-fed mice, db/db and ob/ob mice, and PCOS rats were significantly higher than those of WT mice or rats. In human studies, we find that circulating GPHB5 levels were significantly higher in women with IR and PCOS. GPHB5 levels were positively correlated with age, BMI, WHR, BP, FBG, 2 h-BG, FIns, 2 h-Ins, TC, LDL-C, HbA1c, and FFA, but negatively correlated with adiponectin. Furthermore, GPHB5 was positively correlated with DHEAS and FAI, while negatively correlated with SHBG, FSH, SHBG and FSH. The increased GPHB5 concentration was related to IR and PCOS. After the treatment of metformin, GLP-1RA (Lira), and TZDs, circulating GPHB5 levels were decreased. CONCLUSIONS Our results reveal that circulating GPHB5 could be a biomarker and potential therapeutic target for IR and PCOS in women.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Endocrinology, Chongqing Red Cross Hospital (People’s Hospital of Jiangbei District), Chongqing, China
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ting Xiang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xuyun Xia
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hongmin Zhang
- Department of Endocrinology, The First People’s Hospital of Chongqing Liang Jiang New Area, Chongqing, China
| | - Shan Geng
- Department of Endocrinology, The People’s Hospital of Dazu, Chongqing, China
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Sheng Qiu
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yirui He
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Rui Liu
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ling Li
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Hua Liu
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, MS, United States
| | - Ke Li
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lili Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zerong Liang
- Department of Endocrinology, Chongqing Red Cross Hospital (People’s Hospital of Jiangbei District), Chongqing, China
- *Correspondence: Zerong Liang, ; Jianguo He,
| | - Jianguo He
- Department of Endocrinology, Chongqing Red Cross Hospital (People’s Hospital of Jiangbei District), Chongqing, China
- *Correspondence: Zerong Liang, ; Jianguo He,
| |
Collapse
|
207
|
Liu S, Zhang Y, Yang F, Gu J, Zhang R, Kuang Y, Mai W, Zheng C, Yu Y, Lu R, Zeng L, Cao H, Long Y. Modified Cangfu Daotan decoction ameliorates polycystic ovary syndrome with insulin resistance via NF-κB/LCN-2 signaling pathway in inflammatory microenvironment. Front Endocrinol (Lausanne) 2022; 13:975724. [PMID: 36440213 PMCID: PMC9686851 DOI: 10.3389/fendo.2022.975724] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022] Open
Abstract
This study explored the possible connection between the insulin resistance-targeting protein adipokine lipocalin-2 (LCN-2) and NF-κB signaling pathway in the inflammatory microenvironment in PCOS-IR model rats to determine the pharmacological mechanism of modified Cangfu Daotan decoction (MCDD) intervention for PCOS-IR. We used a high-fat diet (42 days) combined with letrozole (1 mg/kg/day, 42 days) to establish a PCOS-IR rat model. From the third week after modeling, the rats were given continuous administration of MCDD (high dose with 31.68 g/kg, medium dose with 15.84 g/kg, and low dose with 7.92 g/kg) for 28 days. Serum, ovarian tissue, liver, and adipose tissue were collected after the last gavage. Enzyme-linked immunosorbent assay, hematoxylin-eosin (HE) staining, Masson staining, qRT-PCR, and Western blot experiments were performed to detect various indicators. Our results showed that MCDD could reduce body weight and abdominal fat weight; restore normal estrous cycle and ovarian function; alleviate fatty liver; regulate HOMA-IR and OGTT index; reduce serum inflammatory factor levels, LCN-2 level, and gene expression; and regulate the insulin signal transduction and NF-κB pathways in PCOS-IR rats. Thus, MCDD may play a role in improving ovarian function in PCOS-IR rats by downregulating NF-κB/LCN-2 proteins and upregulating the gene expression of Insr/Irs-1/Glut4 in the insulin signaling pathway in the inflammatory environment.
Collapse
Affiliation(s)
- Shuowen Liu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yao Zhang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Fang Yang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jingna Gu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ruyue Zhang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yingying Kuang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wantong Mai
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chengbo Zheng
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yang Yu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ruling Lu
- Department of Gynecology, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Zeng
- Department of Gynecology, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongying Cao
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yongling Long
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- *Correspondence: Yongling Long,
| |
Collapse
|
208
|
Gu Y, Zhou G, Zhou F, Wu Q, Ma C, Zhang Y, Ding J, Hua K. Life Modifications and PCOS: Old Story But New Tales. Front Endocrinol (Lausanne) 2022; 13:808898. [PMID: 35498415 PMCID: PMC9045543 DOI: 10.3389/fendo.2022.808898] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/01/2022] [Indexed: 11/13/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is defined as a kind of endocrine and metabolic disorder that affects female individuals of reproductive age. Lifestyle modifications, including diet modifications, exercise, and behavioral modification, appear to alleviate the metabolic dysfunction and improve the reproductive disorders of PCOS patients (particularly in obese women). Therefore, lifestyle modifications have been gradually acknowledged as the first-line management for PCOS, especially in obese patients with PCOS. However, the mechanism of lifestyle modifications in PCOS, the appropriate composition of diet modifications, and the applicable type of exercise modifications for specific female populations are rarely reported. We conducted a systematic review and enrolled 10 randomized controlled trials for inclusion in a certain selection. In this review, we summarized the existing research on lifestyle modifications in PCOS. We aimed to illustrate the relationship between lifestyle modifications and PCOS (referring to hyperandrogenism, insulin resistance as well as obesity) and also considered the priorities for future research. These results might be an invaluable tool to serve as a guide in lifestyle modifications as the intervention for PCOS and other related endocrine disorders.
Collapse
Affiliation(s)
- Yuanyuan Gu
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Guannan Zhou
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Fangyue Zhou
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiongwei Wu
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
| | - Chengbin Ma
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
| | - Yi Zhang
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
- *Correspondence: Yi Zhang, ; Jingxin Ding, ; Keqin Hua,
| | - Jingxin Ding
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- *Correspondence: Yi Zhang, ; Jingxin Ding, ; Keqin Hua,
| | - Keqin Hua
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- *Correspondence: Yi Zhang, ; Jingxin Ding, ; Keqin Hua,
| |
Collapse
|
209
|
Reproductive outcomes after bariatric surgery in women. Wien Klin Wochenschr 2021; 134:56-62. [PMID: 34878586 PMCID: PMC8813708 DOI: 10.1007/s00508-021-01986-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 01/11/2023]
Abstract
The presence of obesity may significantly influence female fertility through various mechanisms. Impairment of the hypothalamic-pituitary-ovarian axis in obese women may induce anovulation and infertility. Obesity may have an effect on women’s spontaneous and assisted conception rates, increased miscarriage rates, premature labor, stillbirth and perinatal risks, and menstrual irregularity. It has been suggested that weight loss improves reproductive outcomes due to fertility amelioration and an improvement in menstrual irregularity and ovulation. It is still not known which weight reduction procedures (changes in lifestyle, pharmacological management or bariatric intervention) result in optimal outcome on infertility. Currently, bariatric surgery is defined as the best available method for the management of obesity and its associated diseases. We have analyzed literature facts about effects of bariatric surgery on the function of the hypothalamic-pituitary-ovarian axis, polycystic ovary syndrome (PCOS), anti-Mullerian hormone (AMH) and sexual dysfunction in obesity and pregnancy in obesity. Immediate positive effects of bariatric surgery are evident at the moment, while for long-term outcomes more prolonged follow-up investigations should be done.
Collapse
|
210
|
Jin C, Zou K, Xu Y, Yang H, Pan J. Elevated plasma pentraxin-3 in polycystic ovary syndrome is associated with hyperandrogenism: a case-control study. BMC Endocr Disord 2021; 21:240. [PMID: 34856980 PMCID: PMC8641145 DOI: 10.1186/s12902-021-00886-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/25/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Pentraxin 3 (PTX3) - a crucial humoral innate immunity component - is related to obesity and cardiovascular complications in women who suffer from polycystic ovary syndrome (PCOS). However, the circulating PTX3 level in PCOS is still debated. In this study, we aimed to evaluate PTX3 plasma levels in PCOS women of childbearing age, and find possible endocrine/metabolic factors that could affect this level. METHODS A total of 360 women were enrolled: 120 PCOS women and 240 body mass index (BMI) matched normally ovulating women. Blood samples were collected on the third day of natural menstrual cycle or from the bleeding after progesterone withdrawal. The PTX3 concentration was measured by immunoassay. RESULTS The PTX3 plasma level was significantly higher in PCOS women compared to controls. There was a positive correlation between PTX3 plasma level and PCOS diagnosis, overweight, cycle length, serum LH to FSH ratio, estradiol, total testosterone (TT) on the third day of menstrual cycle, antral follicle count (AFC), as well as uric acid. Multivariant linear regression analysis indicated that participants' serum PTX3 levels were proportional to the circulating TT level, existence of PCOS, basal estradiol level and AFC. CONCLUSIONS Overall, the circulating PTX3 level was elevated in PCOS women and significantly associated with the presence of hyperandrogenism. This study provided the basis for further in-depth researches regarding PTX3 role in PCOS pathophysiology.
Collapse
Affiliation(s)
- Congcong Jin
- The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Kexin Zou
- Obstetrics & Gynecology Hospital, Institute of Reproduction and Development, Fudan University, 200011, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, 200030, Shanghai, China
| | - Yue Xu
- Obstetrics & Gynecology Hospital, Institute of Reproduction and Development, Fudan University, 200011, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, 200030, Shanghai, China
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 200011, Shanghai, China
| | - Haiyan Yang
- The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Jiexue Pan
- Obstetrics & Gynecology Hospital, Institute of Reproduction and Development, Fudan University, 200011, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases, 200030, Shanghai, China.
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 200011, Shanghai, China.
| |
Collapse
|
211
|
Yang T, Zhao J, Zhang Q, Liu D, Liu N, Li Y, Yao Z, Zhang Y, Tian F, Liao T, Tang H, Li Y. Associations between dyslipidaemia and pregnancy outcomes in the first complete cycle of IVF/ICSI: a real-world analysis. Reprod Biomed Online 2021; 43:1095-1105. [PMID: 34764017 DOI: 10.1016/j.rbmo.2021.08.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/16/2021] [Accepted: 08/27/2021] [Indexed: 10/20/2022]
Abstract
RESEARCH QUESTION Are there associations between dyslipidaemia and pregnancy outcomes in the first complete cycle of IVF/intracytoplasmic sperm injection (ICSI)? DESIGN This long-term, retrospective real-world analysis involved 5030 infertile women who underwent a first complete IVF/ICSI cycle between January 2015 and October 2020. They were categorized into dyslipidaemia (n = 1903) and control (n = 3127) groups according to serum lipid concentrations before ovarian stimulation. Propensity score matching and multivariable logistic regression were used to control for confounding variables. RESULTS In the raw cohort, women with dyslipidaemia had a significantly increased late miscarriage rate (P = 0.039), decreased term birth rate (P = 0.002) and decreased live birth rate (P = 0.005) compared with non-dyslipidaemic women. In the propensity score-matched cohort, the term birth rate (P = 0.038) and live birth rate (P = 0.044) were significantly lower in the dyslipidaemia group (n = 1686) than the controls (n = 1686). Multivariable logistic regression indicated that infertile women with dyslipidaemia (P = 0.026) and elevated serum total cholesterol concentrations (total cholesterol ≥5.20 mmol/l; P = 0.028) were significantly less likely to have a live birth. Rates of late miscarriage (P = 0.027), term birth (P = 0.003) and live birth (P = 0.010) differed significantly among women with normal, borderline increased and increased serum lipid concentrations. Compared with controls, women with increased serum lipid concentrations had a significantly higher late miscarriage rate, lower term birth rate and lower live birth rate. Women with increased serum lipid concentrations were significantly less likely than controls to have a live birth. CONCLUSIONS Dyslipidaemia, total cholesterol ≥5.20 mmol/l and degrees of elevated serum lipid concentrations are negatively associated with live birth rate in the first complete IVF/ICSI cycle in infertile women.
Collapse
Affiliation(s)
- Tianli Yang
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China
| | - Jing Zhao
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China
| | - Qiong Zhang
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China
| | - Donge Liu
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China
| | - Nenghui Liu
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China
| | - Yumei Li
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China
| | - Zhongyuan Yao
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China
| | - Yeqing Zhang
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China
| | - Fen Tian
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China
| | - Tingting Liao
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China
| | - Hongying Tang
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China.
| | - Yanping Li
- Reproductive Medicine Center, Xiangya Hospital of Central South University, Changsha Hunan, P.R. China; Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha Hunan, P.R. China.
| |
Collapse
|
212
|
Kim KW. Unravelling Polycystic Ovary Syndrome and Its Comorbidities. J Obes Metab Syndr 2021; 30:209-221. [PMID: 34497157 PMCID: PMC8526288 DOI: 10.7570/jomes21043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/11/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a chronic multisystem endocrine disorder that affects women of reproductive age. In the ovary, the dynamic balance between dormant and growing follicles that culminates in ovulation becomes dysfunctional in the presence of excessive androgen production (ovarian/adrenal/peripheral). Moreover, hyperandrogenicity in pregnancy affects fetal development in utero and is linked to maternal pregnancy complications. Hormonal imbalance, ovarian dysfunction, and central obesity often emerge in these patients during adolescence. Once disordered physiological changes develop in PCOS, a vicious cycle ensues, leading to reproductive, metabolic, and psychological comorbidities. With the alarming increase of the number of young adults with a high degree of obesity in Korea, the prevalence of PCOS has also considerably increased. Timely and accurate screening, multicomponent healthy lifestyle modifications for both patients and family members, and comprehensive medical interventions based on international evidence-based guidelines are essential for curtailing PCOS and its comorbidities.
Collapse
Affiliation(s)
- Kyung-Wook Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Dongtan Jeil Women's Hospital and Sangwoon Medical Institute, Hwaseong, Korea.,Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
213
|
Wang Z, Groen H, Cantineau AEP, van Elten TM, Karsten MDA, van Oers AM, Mol BWJ, Roseboom TJ, Hoek A. Effectiveness of a 6-Month Lifestyle Intervention on Diet, Physical Activity, Quality of Life, and Markers of Cardiometabolic Health in Women with PCOS and Obesity and Non-PCOS Obese Controls: One Size Fits All? Nutrients 2021; 13:nu13103425. [PMID: 34684438 PMCID: PMC8538637 DOI: 10.3390/nu13103425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 12/27/2022] Open
Abstract
Little is known about the difference in effectiveness of lifestyle intervention between women with PCOS and non-PCOS women. In a post hoc longitudinal analysis of a randomized, controlled trial, we aimed to investigate whether infertile women with PCOS and obesity (N = 87) responded differently to a 6-month lifestyle intervention program than infertile non-PCOS obese controls (N = 172). We evaluated several aspects of the intervention such as changes in diet, physical activity, and dropout rate, as well as the effect on weight, quality of life (QoL), and cardiometabolic outcomes. Multilevel analyses were used, and analyses were adjusted for baseline characteristics such as age, education, and smoking. Although BMI in both groups significantly decreased at 3 months and 6 months, there were no significant differences between the groups at 3 months (adjusted B: −0.3, 95% CI: −0.9 to 0.3, p = 0.35) and 6 months (adjusted B: 0.5, 95% CI: −0.4 to 1.4, p = 0.29). Women with PCOS and non-PCOS women had similar compliance with the lifestyle intervention in terms of actual change in diet and physical activity. Mental QoL scores were not different at either 3 or 6 months. Physical QoL scores were lower in women with PCOS compared with non-PCOS women at 3 months (adjusted B: −2.4, 95% CI: −4.8 to −0.06, p = 0.045) but not at 6 months. Cardiometabolic parameters did not differ between the groups. Our results showed that infertile women with PCOS and obesity and non-PCOS obese controls responded largely similarly to our lifestyle intervention and achieved the same level of improvement in markers of cardiometabolic health.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Centre Groningen, 9700 RB Groningen, The Netherlands; (Z.W.); (A.E.P.C.); (A.M.v.O.)
| | - Henk Groen
- Department of Epidemiology, University of Groningen, University Medical Centre Groningen, 9700 RB Groningen, The Netherlands;
| | - Astrid E. P. Cantineau
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Centre Groningen, 9700 RB Groningen, The Netherlands; (Z.W.); (A.E.P.C.); (A.M.v.O.)
| | - Tessa M. van Elten
- Department of Public and Occupational Health, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1105 AZ Amsterdam, The Netherlands;
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.D.A.K.); (T.J.R.)
- Department of Obstetrics and Gynecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, 1105 AZ Amsterdam, The Netherlands
| | - Matty D. A. Karsten
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.D.A.K.); (T.J.R.)
- Department of Obstetrics and Gynecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, 1105 AZ Amsterdam, The Netherlands
| | - Anne M. van Oers
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Centre Groningen, 9700 RB Groningen, The Netherlands; (Z.W.); (A.E.P.C.); (A.M.v.O.)
| | - Ben W. J. Mol
- Department of Obstetrics and Gynecology, Monash University, Melbourne 3800, Australia;
| | - Tessa J. Roseboom
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.D.A.K.); (T.J.R.)
- Department of Obstetrics and Gynecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, 1105 AZ Amsterdam, The Netherlands
| | - Annemieke Hoek
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Centre Groningen, 9700 RB Groningen, The Netherlands; (Z.W.); (A.E.P.C.); (A.M.v.O.)
- Correspondence:
| |
Collapse
|
214
|
Involvement of Kisspeptin in androgen-induced hypothalamic endoplasmic reticulum stress and its rescuing effect in PCOS rats. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166242. [PMID: 34389474 DOI: 10.1016/j.bbadis.2021.166242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 11/24/2022]
Abstract
Endoplasmic reticulum (ER) stress, with adaptive unfolded protein response (UPR), is a key link between obesity, insulin resistance and type 2 diabetes, all of which are often present in the most common endocrine-metabolic disorder in women of reproductive age, polycystic ovary syndrome (PCOS), which is characterized with hyperandrogenism. However, the link between excess androgen and Endoplasmic reticulum (ER) stress/insulin resistance in patients with polycystic ovary syndrome (PCOS) is unknown. An unexpected role of kisspeptin was reported in the regulation of UPR pathways and its involvement in the androgen-induced ER stress in hypothalamic neuronal cells. To evaluate the relationship of kisspeptin and ER stress, we detected Kisspeptin and other factors in blood plasm of PCOS patients, rat models and hypothalamic neuronal cells. We detected higher testosterone and lower kisspeptin levels in the plasma of PCOS than that in non-PCOS women. We established a PCOS rat model by dihydrotestosterone (DHT) chronic exposure, and observed significantly downregulated kisspeptin expression and activated UPR pathways in PCOS rat hypothalamus compared to that in controls. Inhibition or knockdown of kisspeptin completely mimicked the enhancing effect of DHT on UPR pathways in a hypothalamic neuronal cell line, GT1-7. Kp10, the most potent peptide of kisspeptin, effectively reversed or suppressed the activated UPR pathways induced by DHT or thapsigargin, an ER stress activator, in GT1-7 cells, as well as in the hypothalamus in PCOS rats. Similarly, Kisspeptin attenuated thapsigargin-induced Ca2+ response and the DHT- induced insulin resistance in GT1-7 cells. Collectively, the present study has revealed an unexpected protective role of kisspeptin against ER stress and insulin resistance in the hypothalamus and provided a new treatment strategy targeting hypothalamic ER stress and insulin resistance with kisspeptin as a potential therapeutic agent.
Collapse
|
215
|
Zhang Y, Hu M, Yang F, Zhang Y, Ma S, Zhang D, Wang X, Sferruzzi-Perri AN, Wu X, Brännström M, Shao LR, Billig H. Increased uterine androgen receptor protein abundance results in implantation and mitochondrial defects in pregnant rats with hyperandrogenism and insulin resistance. J Mol Med (Berl) 2021; 99:1427-1446. [PMID: 34180022 PMCID: PMC8455403 DOI: 10.1007/s00109-021-02104-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/20/2021] [Accepted: 06/10/2021] [Indexed: 12/21/2022]
Abstract
Abstract In this study, we show that during normal rat pregnancy, there is a gestational stage-dependent decrease in androgen receptor (AR) abundance in the gravid uterus and that this is correlated with the differential expression of endometrial receptivity and decidualization genes during early and mid-gestation. In contrast, exposure to 5α-dihydrotestosterone (DHT) and insulin (INS) or DHT alone significantly increased AR protein levels in the uterus in association with the aberrant expression of endometrial receptivity and decidualization genes, as well as disrupted implantation. Next, we assessed the functional relevance of the androgen-AR axis in the uterus for reproductive outcomes by treating normal pregnant rats and pregnant rats exposed to DHT and INS with the anti-androgen flutamide. We found that AR blockage using flutamide largely attenuated the DHT and INS-induced maternal endocrine, metabolic, and fertility impairments in pregnant rats in association with suppressed induction of uterine AR protein abundance and androgen-regulated response protein and normalized expression of several endometrial receptivity and decidualization genes. Further, blockade of AR normalized the expression of the mitochondrial biogenesis marker Nrf1 and the mitochondrial functional proteins Complexes I and II, VDAC, and PHB1. However, flutamide treatment did not rescue the compromised mitochondrial structure resulting from co-exposure to DHT and INS. These results demonstrate that functional AR protein is an important factor for gravid uterine function. Impairments in the uterine androgen-AR axis are accompanied by decreased endometrial receptivity, decidualization, and mitochondrial dysfunction, which might contribute to abnormal implantation in pregnant PCOS patients with compromised pregnancy outcomes and subfertility. Key messages The proper regulation of uterine androgen receptor (AR) contributes to a
normal pregnancy process, whereas the aberrant regulation of uterine AR might
be linked to polycystic ovary syndrome (PCOS)-induced pregnancy-related
complications. In the current study, we found that during normal rat pregnancy there is
a stage-dependent decrease in AR abundance in the gravid uterus and that this
is correlated with the differential expression of the endometrial receptivity
and decidualization genes Spp1, Prl, Igfbp1,
and Hbegf. Pregnant rats exposed to 5α-dihydrotestosterone (DHT) and insulin (INS)
or to DHT alone show elevated uterine AR protein abundance and implantation
failure related to the aberrant expression of genes involved in endometrial
receptivity and decidualization in early to mid-gestation. Treatment with the anti-androgen flutamide, starting from
pre-implantation, effectively prevents DHT + INS-induced defects in endometrial
receptivity and decidualization gene expression, restores uterine mitochondrial
homeostasis, and increases the pregnancy rate and the numbers of viable
fetuses. This study adds to our understanding of the mechanisms underlying poor
pregnancy outcomes in PCOS patients and the possible therapeutic use of
anti-androgens, including flutamide, after spontaneous conception.
Supplementary Information The online version contains supplementary material available at 10.1007/s00109-021-02104-z.
Collapse
Affiliation(s)
- Yuehui Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.,Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 11, P. O. Box 434, 40530, Gothenburg, Sweden
| | - Min Hu
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 11, P. O. Box 434, 40530, Gothenburg, Sweden.,Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.,Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, 510120, China
| | - Fan Yang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yizhuo Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Shuting Ma
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Dongqi Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xu Wang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Amanda Nancy Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Xiaoke Wu
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Gothenburg, 41345, Gothenburg, Sweden
| | - Linus R Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 11, P. O. Box 434, 40530, Gothenburg, Sweden.
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 11, P. O. Box 434, 40530, Gothenburg, Sweden
| |
Collapse
|
216
|
Ali ZE, Uraji J, Serdarogullari M, Sonnen KF, Celik-Ozenci C, Macklon N, Massarotti C. #ESHREjc report: are fallopian tubes the unsung hero of reproduction? Hum Reprod 2021; 36:1728-1729. [PMID: 34002213 DOI: 10.1093/humrep/deab109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Julia Uraji
- IVF Laboratory, MVZ VivaNeo Kinderwunschzentrum, Düsseldorf, Germany
| | - Munevver Serdarogullari
- Department of Histology and Embryology, Faculty of Medicine, Cyprus International University, Çukurova/Adana, Turkey
| | - Katharina F Sonnen
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ciler Celik-Ozenci
- Department of Histology and Embryology, Akdeniz University Medical Faculty, Antalya, Turkey
| | - Nicholas Macklon
- London Women's Clinic, London, UK.,Department of Gynaecology, Obstetrics and Fertility, Zealand University Hospital, Køge, Denmark
| | - Claudia Massarotti
- Academic Unit of Obstetrics and Gynecology, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy
| |
Collapse
|
217
|
Zhou G, Gu Y, Zhou F, Zhang M, Zhang G, Wu L, Hua K, Ding J. The Emerging Roles and Therapeutic Potential of Extracellular Vesicles in Infertility. Front Endocrinol (Lausanne) 2021; 12:758206. [PMID: 34745016 PMCID: PMC8569852 DOI: 10.3389/fendo.2021.758206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/28/2021] [Indexed: 12/20/2022] Open
Abstract
Infertility is becoming much more common and affects more couples. The past years witnessed the rapid development of the diagnosis and treatment upon infertility, which give numerous coupled more opportunities become parents. Extracellular vesicles are known as nano-sized membrane vesicles to play a major role in intracellular communication. In recent years, several basic and clinical studies have tried to investigate the correlation between the reproductive health/disorder and extracellular vesicles. However, the mechanism is still unclear. In this review, we reviewed the relationship between reproductive physiology and extracellular vesicles, and then collectively focused on the recent findings on the relationship between extracellular and infertility, and its consequent influence on the novel insight regarding the therapeutic strategies for infertility in the future clinical practice.
Collapse
Affiliation(s)
- Guannan Zhou
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yuanyuan Gu
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
| | - Fangyue Zhou
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Menglei Zhang
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Ganrong Zhang
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Ligang Wu
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Jingxin Ding, ; Keqin Hua, ; Ligang Wu,
| | - Keqin Hua
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- *Correspondence: Jingxin Ding, ; Keqin Hua, ; Ligang Wu,
| | - Jingxin Ding
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- *Correspondence: Jingxin Ding, ; Keqin Hua, ; Ligang Wu,
| |
Collapse
|
218
|
Lu X, Huang L, Huang Z, Feng D, Clark RJ, Chen C. LEAP-2: An Emerging Endogenous Ghrelin Receptor Antagonist in the Pathophysiology of Obesity. Front Endocrinol (Lausanne) 2021; 12:717544. [PMID: 34512549 PMCID: PMC8428150 DOI: 10.3389/fendo.2021.717544] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/28/2021] [Indexed: 11/20/2022] Open
Abstract
Liver-expressed antimicrobial peptide 2 (LEAP-2), originally described as an antimicrobial peptide, has recently been recognized as an endogenous blocker of growth hormone secretagogue receptor 1a (GHS-R1a). GHS-R1a, also known as ghrelin receptor, is a G protein-coupled receptor (GPCR) widely distributed on the hypothalamus and pituitary gland where it exerts its major functions of regulating appetite and growth hormone (GH) secretion. The activity of GHS-R1a is controlled by two counter-regulatory endogenous ligands: Ghrelin (activation) and LEAP-2 (inhibition). Ghrelin activates GHS-R1a on the neuropeptide Y/Agouti-related protein (NPY/AgRP) neurons at the arcuate nucleus (ARC) to promote appetite, and on the pituitary somatotrophs to stimulate GH release. On the flip side, LEAP-2, acts both as an endogenous competitive antagonist of ghrelin and an inverse agonist of constitutive GHS-R1a activity. Such a biological property of LEAP-2 vigorously blocks ghrelin's effects on food intake and hormonal secretion. In circulation, LEAP-2 displays an inverse pattern as to ghrelin; it increases with food intake and obesity (positive energy balance), whereas decreases upon fasting and weight loss (negative energy balance). Thus, the LEAP-2/ghrelin molar ratio fluctuates in response to energy status and modulation of this ratio conversely influences energy intake. Inhibiting ghrelin's activity has shown beneficial effects on obesity in preclinical experiments, which sheds light on LEAP-2's anti-obesity potential. In this review, we will analyze LEAP-2's effects from a metabolic point of view with a focus on metabolic hormones (e.g., ghrelin, GH, and insulin), and discuss LEAP-2's potential as a promising therapeutic target for obesity.
Collapse
Affiliation(s)
- Xuehan Lu
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Lili Huang
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Zhengxiang Huang
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Dandan Feng
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
- Department of Physiology, Xiangya Medical School, Central South University, Changsha, China
| | - Richard J. Clark
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
- *Correspondence: Chen Chen,
| |
Collapse
|
219
|
Jiang Q, Pan Y, Li P, Zheng Y, Bian Y, Wang W, Wu G, Song T, Shi Y. ANGPTL4 Expression in Ovarian Granulosa Cells Is Associated With Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2021; 12:799833. [PMID: 35140683 PMCID: PMC8820586 DOI: 10.3389/fendo.2021.799833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES This study aims to characterize the expression of ANGPTL4 in ovarian granulosa cells (GCs) and its association with polycystic ovary syndrome (PCOS). METHODS This study included 104 PCOS patients and 112 women in control group undergoing in vitro fertilization-embryo transfer (IVF-ET) from the reproductive hospital affiliated with Shandong University from 2019 to 2021. By reverse transcription and real-time quantitative (RT-q) PCR, the mRNA expression of ANGPTL4 in GCs was assessed, and clinical information for these patients were then reviewed and analyzed. RESULTS The RT-qPCR results showed that ANGPTL4 expression in the control group was significantly lower than that in the PCOS group (p = 0.000) and had positive association with AMH (r = 0.211), HOMA-IR (r = 0.174), LDL/HDL (r = 0.176), ApoB/ApoAI (r = 0.155), and TC/HDL (r = 0.189). Additionally, the high expression of ANGPTL4 in the ovarian granulosa cells might be an independent predictor in PCOS (OR: 3.345; 95% CI: 1.951-5.734) with a close contact with incidence of PCOS (AUC: 0.704; 95% CI: 0.633-0.774, p < 0.001). CONCLUSIONS Our study revealed higher ANGPTL4 expression in ovarian GCs with PCOS. Its association with glucose and lipid metabolism showed that ANGPTL4 might play an important role in PCOS metabolism and pathogenesis.
Collapse
Affiliation(s)
- Qi Jiang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Ye Pan
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Ping Li
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yanjun Zheng
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Yuehong Bian
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Wenqi Wang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Guihua Wu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tian Song
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Yuhua Shi
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- *Correspondence: Yuhua Shi,
| |
Collapse
|