1
|
Peng H, Ren J, Zhao Y, Fang X, Wang X, Liu C, Wan Z. Unraveling the Connection between PCOS and renal Complications: Current insights and Future Directions. Diabetes Res Clin Pract 2025:112235. [PMID: 40334925 DOI: 10.1016/j.diabres.2025.112235] [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: 03/11/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/09/2025]
Abstract
Polycystic ovary syndrome (PCOS) represents the most prevalent endocrine disorder among women of reproductive age, affecting approximately 5-18% of females worldwide. Characterized by irregular ovulation, hyperandrogenism, and polycystic ovaries, hyperandrogenism is the defining feature. Recent evidence highlights that, in addition to its notable reproductive and metabolic consequences, PCOS may also contribute to an elevated risk of renal complications. This increased risk is attributed to chronic low-grade inflammation, hormonal dysregulation, and disturbances in lipid metabolism inherent to the condition. However, the pathological mechanisms, clinical manifestations, and progression of secondary renal damage in this cohort remain insufficiently studied. This review consolidates current understanding of the relationship between PCOS and chronic kidney disease (CKD), aiming to clarify potential mechanisms by which PCOS may induce secondary renal dysfunction, encompassing both direct renal impairment and indirect damage mediated through systemic alterations. Furthermore, it advocates for comprehensive management strategies to mitigate renal risks in patients with PCOS, emphasizing the necessity of multidisciplinary approaches and further research to address these critical gaps.
Collapse
Affiliation(s)
- Haoyu Peng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Junyi Ren
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Zhao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinyi Fang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoxiao Wang
- Department of Organ Transplantation, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chi Liu
- Department of Nephrology, Sichuan Clinical Research Center for Kidney Disease, Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, China.
| | - Zhengwei Wan
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
2
|
Meng Y, Zhao T, Zhang R, Zhu X, Ma C, Shi Q. Global burden of polycystic ovary syndrome among women of childbearing age, 1990-2021: a systematic analysis using the global burden of disease study 2021. Front Public Health 2025; 13:1514250. [PMID: 40206176 PMCID: PMC11979288 DOI: 10.3389/fpubh.2025.1514250] [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: 10/22/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND The escalating global incidence of polycystic ovary syndrome (PCOS) necessitates a thorough examination of its epidemiological trends and sociodemographic correlations. Our study bridges this gap by analyzing the global impact of PCOS among women of childbearing age (WCBA) from 1990 to 2021, aiming to inform strategies for enhanced prevention and management. METHODS We extracted data from the Global Burden of Disease Study 2021 (GBD 2021), focusing on the burden of PCOS among women aged 15-49 years. We assessed incidence, prevalence, and disability-adjusted life years (DALYs) trends using the estimated annual percentage change (EAPC) and explored the link between PCOS burden and sociodemographic index (SDI). RESULTS In 2021, the prevalence cases of PCOS worldwide were 65.77 million, the incidence cases were 1,175.07 thousand, and the DALYs cases were 576.05 thousand. Compared with 1990, the percentage changes were 89, 49, and 87%, respectively. The EAPCs indicated upward trends in prevalence and DALYs rates, with a less pronounced increase in incidence rates. The middle SDI region had the highest PCOS case numbers, and the 45-49 age group in this region experienced the most significant burden increase. A strong positive correlation existed between PCOS prevalent rates and SDI (r = 0.582, p < 0.001). CONCLUSION The global burden of PCOS among WCBA has significantly increased over the past three decades, particularly in the 45-49 age group in middle SDI regions. The correlation between PCOS prevalent rates and SDI highlights the role of socio-economic factors in PCOS epidemiology. Tailored prevention and management strategies are crucial for reducing the global burden of PCOS and improving female health outcomes.
Collapse
Affiliation(s)
- Yaping Meng
- Department of Obstetrics and Gynecology, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Ting Zhao
- Department of Obstetrics and Gynecology, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Rui Zhang
- Department of Obstetrics and Gynecology, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xiaoyan Zhu
- Department of Obstetrics and Gynecology, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Chao Ma
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Qin Shi
- Department of Obstetrics and Gynecology, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
3
|
Senior AM, Raubenheimer D, Couteur DGL, Simpson SJ. The Geometric Framework for Nutrition and Its Application to Rodent Models. Annu Rev Anim Biosci 2025; 13:389-410. [PMID: 39546416 DOI: 10.1146/annurev-animal-111523-102327] [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: 11/17/2024]
Abstract
Rodents have been the primary model for mammalian nutritional physiology for decades. Despite an extensive body of literature, controversies remain around the effects of specific nutrients and total energy intake on several aspects of nutritional biology, even in this well-studied model. One approach that is helping to bring clarity to the field is the geometric framework for nutrition (GFN). The GFN is a multidimensional paradigm that can be used to conceptualize nutrition and nutritional effects, design experiments, and interpret results. To date, more than 30 publications have applied the GFN to data from rodent models of nutrition. Here we review the major conclusions from these studies. We pay particular attention to the effects of macronutrients on satiety, glucose metabolism, lifespan and the biology of aging, reproductive function, immune function, and the microbiome. We finish by highlighting several knowledge gaps that became evident upon reviewing this literature.
Collapse
Affiliation(s)
- Alistair M Senior
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, New South Wales, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| | - David Raubenheimer
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| | - David G Le Couteur
- ANZAC Research Institute, The Concord Hospital, Concord, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| | - Stephen J Simpson
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| |
Collapse
|
4
|
Ahmad F, Ahmed SH, Choucair F, Chouliaras S, Awwad J, Terranegra A. A disturbed communication between hypothalamic-pituitary-ovary axis and gut microbiota in female infertility: is diet to blame? J Transl Med 2025; 23:92. [PMID: 39838491 PMCID: PMC11749209 DOI: 10.1186/s12967-025-06117-x] [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: 11/28/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
Female infertility is a multifactorial condition influenced by various genetic, environmental, and lifestyle factors. Recent research has investigated the significant impact of gut microbiome dysbiosis on systemic inflammation, metabolic dysfunction, and hormonal imbalances, which can potentially impair fertility. The gut-brain axis, a bidirectional communication system between the gut and the brain, also plays a significant role in regulating reproductive functions. Emerging evidence suggests that the gut microbiome can influence brain functions and behavior, further emphasizing the importance of the microbiota-gut-brain axis in reproduction. Given their role as a major modulator of the gut microbiome, diet and dietary factors, including dietary patterns and nutrient intake, have been implicated in the development and management of female infertility. Hence, this review aims to highlight the impact of dietary patterns, such as the Western diet (WD) and Mediterranean diet (MD), and to decipher their modulatory action on the microbiota-gut-brain axis in infertile women. By contrasting the detrimental effects of WD with the therapeutic potential of MD, we emphasize the pivotal role of a balanced diet rich in nutrients in promoting a healthy gut microbiome. These insights underscore the potential of targeted dietary interventions and lifestyle modifications as promising strategies to enhance reproductive outcomes in subfertile women.
Collapse
Affiliation(s)
- Fatima Ahmad
- Translational Medicine Department, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar
| | - Salma H Ahmed
- Translational Medicine Department, Sidra Medicine, Doha, Qatar
| | - Fadi Choucair
- Reproductive Medicine Unit, Sidra Medicine, Doha, Qatar
| | - Spyridon Chouliaras
- Reproductive Medicine Unit, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine, Ar-Rayyan, Qatar
| | - Johnny Awwad
- Reproductive Medicine Unit, Sidra Medicine, Doha, Qatar
- Vincent Memorial Obstetrics and Gynecology Service, Massachusetts General Hospital, Boston, MA, USA
| | - Annalisa Terranegra
- Translational Medicine Department, Sidra Medicine, Doha, Qatar.
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar.
| |
Collapse
|
5
|
Dilliyappan S, Kumar AS, Venkatesalu S, Palaniyandi T, Baskar G, Sivaji A, Rab SO, Saeed M, Shivaranjani KS. Polycystic ovary syndrome: Recent research and therapeutic advancements. Life Sci 2024; 359:123221. [PMID: 39521272 DOI: 10.1016/j.lfs.2024.123221] [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: 07/01/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Polycystic ovary syndrome is often characterized by the appearance of several tiny cysts (fluid-filled sacs) in the ovaries. It is the most significant endocrinopathy affecting 8-13 % of women during their lifetime. Within the dynamic domain of women's health, this syndrome is a widespread issue that presents with an array of signs, including insulin resistance, hirsutism, androgen development, and menstrual flaws prompted by genetic, diet/lifestyle, gut microbiota dysbiosis, and environmental toxins. Impaired folliculogenesis, aberrant cortisol metabolism, and genes associated with steroidogenesis contribute to the pathophysiology of the disease. Moreover, it combines with various concurrent metabolic and idiopathic conditions specifically type 2 diabetes, heart disease, cancer, and infertility. On persuading the reproductive framework of women from ontogeny to menopause, the complexity of the syndrome hereditates generations due to maternal inheritance of hyperandrogenism. The advancement in diagnostic norms paved the way from the Rotterdam criteria to metabolomics, 3D ultrasound, and assisted reproductive technologies. The management and treatment of this hormonal disorder can be prevailed through lifestyle modifications and prompt medications. This review entails the aforementioned benchmarks of the syndrome's complexity and its ongoing research in alleviating its intricate behavioral changes in women from in-utero to menopause.
Collapse
Affiliation(s)
| | - Avanthika Satish Kumar
- Department of Biotechnology, Dr.M.G.R. Educational and Research Institute, Chennai, India
| | - Sneha Venkatesalu
- Department of Biotechnology, Dr.M.G.R. Educational and Research Institute, Chennai, India
| | - Thirunavukkarasu Palaniyandi
- Department of Biotechnology, Dr.M.G.R. Educational and Research Institute, Chennai, India; ACS-Advanced Medical Research Institute, Dr. M.G.R Educational and Research Institute, Maduravoyal, Chennai 600095, India.
| | - Gomathy Baskar
- Department of Biotechnology, Dr.M.G.R. Educational and Research Institute, Chennai, India
| | - Asha Sivaji
- Department of Biochemistry, DKM College for Women, Vellore, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - K S Shivaranjani
- Department of Gynecology, Sri Lalithambigai Medical College and Hospital, Chennai, India
| |
Collapse
|
6
|
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
|
7
|
Chen P, Ni S, Liu QF, Ou-Yang L. Retinol intake and PCOS management: a plasma metabolite and protein analysis via Mendelian randomization and NHANES 2011-2016. Front Nutr 2024; 11:1434648. [PMID: 39507894 PMCID: PMC11537956 DOI: 10.3389/fnut.2024.1434648] [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: 05/18/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Background Polycystic Ovary Syndrome (PCOS) represents a complex endocrine disorder characterized by a significant interplay with metabolic dysfunction and obesity. This research endeavors to elucidate the causal dynamics among plasma metabolites, proteins, and PCOS, alongside Body Mass Index (BMI), to pinpoint prospective therapeutic interventions. Methods This investigation employed Mendelian randomization (MR) analyses combined with data derived from the National Health and Nutrition Examination Survey (NHANES) to explore the relationships between 1,400 plasma metabolites and PCOS, factoring in BMI adjustments. Additionally, the study examined the influence of plasma proteins and performed a retrospective cross-sectional analysis focusing on retinol consumption and testosterone levels. Results MR analyses showed metabolite Glycosyl-N-(2-hydroxynervonoyl)-sphingosine (GNS) and protein Keratin 19 (KRT19) were identified as significant markers in the context of PCOS and BMI adjustments. A Phenome-Wide Association Study (PheWAS) underscored the linkage between KRT19 and BMI, while gene-drug interaction findings demonstrated a connection between KRT19 and retinol. Analysis for NHANES data disclosed a negative correlation between retinol intake and testosterone levels, particularly within normal weight and obese cohorts, suggesting the feasibility of dietary interventions for PCOS management. Conclusion The study sheds light on the intricate interactions between plasma metabolites, proteins, and PCOS, considering BMI variations, and highlights KRT19 protein as a promising therapeutic target. The outcomes support the integration of retinol consumption into dietary strategies to regulate testosterone levels and potentially alleviate PCOS symptoms, underscoring the necessity for personalized nutritional and therapeutic approaches in the effective management of PCOS.
Collapse
Affiliation(s)
| | | | - Qi-Fang Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | | |
Collapse
|
8
|
Guan HR, Li B, Zhang ZH, Wu HS, Wang N, Chen XF, Zhou CL, Bian XR, Li L, Xu WF, He XL, Dong YJ, Jiang NH, Su J, Lv GY, Chen SH. Exploring the efficacy and mechanism of Bailing capsule to improve polycystic ovary syndrome in mice based on intestinal-derived LPS-TLR4 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118274. [PMID: 38697410 DOI: 10.1016/j.jep.2024.118274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/20/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Polycystic ovary syndrome (PCOS) is a common endocrine disorder associated with reproductive dysfunction and metabolic abnormalities, particularly characterized by insulin resistance and chronic low-grade inflammation. Multiple clinical studies have clearly demonstrated the significant efficacy and safety of the combination of Bailing capsules (BL) in the treatment of PCOS, but its pharmacological effects and mechanisms still require further study. AIM OF THE STUDY To evaluate the effect of BL on improving PCOS in mice and explore the mechanism. METHODS In this study, Dehydroepiandrosterone (DHEA) injection was administered alone and in combination with a high-fat and high-sugar diet to induce PCOS-like mouse. They were randomly divided into five groups: normal group (N), PCOS group (P), Bailing capsule low-dose group (BL-L), Bailing capsule high-dose group (BL-H) and Metformin + Daine-35 group (M + D). Firstly, the effects of BL on ovarian lesions, serum hormone levels, HOMA-IR, intestinal barrier function, inflammation levels, along with the expression of IRS1, PI3K, AKT, TLR4, Myd88, NF-κB p65, TNF-α, IL-6, and Occludin of the ovary, liver and colon were investigated. Finally, the composition of the gut microbiome of fecal was tested. RESULTS The administration of BL significantly reduced body weight, improved hormone levels, improved IR, and attenuated pathological damage to ovarian tissues, up-regulated the expression of IRS1, PI3K, and AKT in liver. It also decreased serum LPS, TNF-α, and IL-6 levels, while downregulating the expression of Myd88, TLR4, and NF-κB p65. Additionally, BL improved intestinal barrier damage and upregulated the expression of Occludin. Interestingly, the abundance of norank_f__Muribaculacea and Lactobacillus was down-regulated, while the abundance of Akkermansia was significantly up-regulated. CONCLUSION The results of the study showed that BL exerts a treatment PCOS effect, which may be related to the modulation of the gut microbiota, the improvement of insulin resistance and the intestinal-derived LPS-TLR4 inflammatory pathway. Our research will provide a theoretical basis for the clinical treatment of PCOS.
Collapse
Affiliation(s)
- Hao-Ru Guan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Bo Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang Province, 313000, PR China
| | - Ze-Hua Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Han-Song Wu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Ning Wang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Xian-Fang Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Cheng-Liang Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Xue-Ren Bian
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Lu Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Wan-Feng Xu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Xing-Lishang He
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Ying-Jie Dong
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Ning-Hua Jiang
- The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, PR China.
| | - Jie Su
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| | - Gui-Yuan Lv
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| | - Su-Hong Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang Province, 313000, PR China.
| |
Collapse
|
9
|
Zhang KH, Zhang FF, Zhang ZL, Fang KF, Sun WX, Kong N, Wu M, Liu HO, Liu Y, Li Z, Cai QQ, Wang Y, Wei QW, Lin PC, Lin Y, Xu W, Xu CJ, Yuan YY, Zhao SM. Follicle stimulating hormone controls granulosa cell glutamine synthesis to regulate ovulation. Protein Cell 2024; 15:512-529. [PMID: 38167949 PMCID: PMC11214834 DOI: 10.1093/procel/pwad065] [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: 08/23/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is the leading cause of anovulatory infertility. Inadequate understanding of the ovulation drivers hinders PCOS intervention. Herein, we report that follicle stimulating hormone (FSH) controls follicular fluid (FF) glutamine levels to determine ovulation. Murine ovulation starts from FF-exposing granulosa cell (GC) apoptosis. FF glutamine, which decreases in pre-ovulation porcine FF, elevates in PCOS patients FF. High-glutamine chow to elevate FF glutamine inhibits mouse GC apoptosis and induces hormonal, metabolic, and morphologic PCOS traits. Mechanistically, follicle-development-driving FSH promotes GC glutamine synthesis to elevate FF glutamine, which maintain follicle wall integrity by inhibiting GC apoptosis through inactivating ASK1-JNK apoptotic pathway. FSH and glutamine inhibit the rapture of cultured murine follicles. Glutamine removal or ASK1-JNK pathway activation with metformin or AT-101 reversed PCOS traits in PCOS models that are induced with either glutamine or EsR1-KO. These suggest that glutamine, FSH, and ASK1-JNK pathway are targetable to alleviate PCOS.
Collapse
Affiliation(s)
- Kai-Hui Zhang
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
- Shanghai Key Laboratory of Metabolic Remodeling, and Children’s Hospital of Fudan University, Shanghai 200032, China
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University (Jinan Children’s Hospital), Jinan 250022, China
| | - Fei-Fei Zhang
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
| | - Zhi-Ling Zhang
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
- Shanghai Key Laboratory of Metabolic Remodeling, and Children’s Hospital of Fudan University, Shanghai 200032, China
- School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Ke-Fei Fang
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
| | - Wen-Xing Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226019, China
| | - Na Kong
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Min Wu
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Hai-Ou Liu
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
| | - Yan Liu
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
| | - Zhi Li
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
| | - Qing-Qing Cai
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
| | - Yang Wang
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
| | - Quan-Wei Wei
- Department of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210014, China
| | - Peng-Cheng Lin
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai University for Nationalities, Xining 810007, China
| | - Yan Lin
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
- Shanghai Key Laboratory of Metabolic Remodeling, and Children’s Hospital of Fudan University, Shanghai 200032, China
| | - Wei Xu
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
- Shanghai Key Laboratory of Metabolic Remodeling, and Children’s Hospital of Fudan University, Shanghai 200032, China
- Shanghai Fifth People’s Hospital of Fudan University, Fudan University, Shanghai 200240, China
| | - Cong-Jian Xu
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
| | - Yi-Yuan Yuan
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
- Shanghai Key Laboratory of Metabolic Remodeling, and Children’s Hospital of Fudan University, Shanghai 200032, China
| | - Shi-Min Zhao
- The Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200090, China
- Shanghai Key Laboratory of Metabolic Remodeling, and Children’s Hospital of Fudan University, Shanghai 200032, China
- School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai University for Nationalities, Xining 810007, China
| |
Collapse
|
10
|
Vadan RL, Varela N, Zhuravko N, Ogidan NO, Adedara VO, Keku E. Comparative Management Methods for Adolescents With Polycystic Ovarian Syndrome: A Systemic Review. Cureus 2024; 16:e55876. [PMID: 38595887 PMCID: PMC11003559 DOI: 10.7759/cureus.55876] [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] [Accepted: 03/09/2024] [Indexed: 04/11/2024] Open
Abstract
Polycystic ovarian syndrome (PCOS) is a common endocrinological disorder affecting many adolescents and women of reproductive age worldwide. A diagnosis of PCOS in adolescence relies upon investigating each medical history independently and noting commonly associated symptoms, including obesity, insulin resistance, acne, menstrual abnormalities, and hirsutism. Many researchers are aiming to discover a methodology to help manage the symptoms associated with PCOS, especially in adolescents. This review will investigate management methods possible for adolescents with PCOS. Although the most preferred way to help reduce symptoms is through lifestyle modifications such as vigorous exercise and dietary regimens low in carbohydrates, pharmaceuticals are also offering promising results to adolescents with PCOS. Metformin, oral contraceptives, gonadotropin-releasing hormone (GnRH) antagonists, and other alternatives, including finasteride, eflornithine, fibroblast growth factors (FGFs), and vitamin D, are all shown to help improve insulin sensitivity and regulate menstrual cycles and reduce hirsutism. Epilatory and surgical measurements are also available; however, they are reserved for when all other methods fail and once adulthood or an appropriate age is reached. Although there are many pharmaceuticals available, it is necessary to evaluate each adolescent with PCOS uniquely and prescribe the appropriate pharmacotherapy regarding their symptoms.
Collapse
Affiliation(s)
- Roberta L Vadan
- Medicine, St. George's University School of Medicine, St. George's, GRD
| | - Nanette Varela
- Medicine, St. George's University School of Medicine, St. George's, GRD
| | - Nikita Zhuravko
- Medicine, St. George's University School of Medicine, St. George's, GRD
| | - Noreena O Ogidan
- Medicine, St. George's University School of Medicine, St. George's, GRD
| | - Victor O Adedara
- Medicine, St. George's University School of Medicine, St. George's, GRD
| | - Emmanuel Keku
- Public Health & Preventive Medicine, St. George's University School of Medicine, St. George's, GRD
| |
Collapse
|
11
|
Xu Q, Zhang J, Lu Y, Wu L. Association of metabolic-dysfunction associated steatotic liver disease with polycystic ovary syndrome. iScience 2024; 27:108783. [PMID: 38292434 PMCID: PMC10825666 DOI: 10.1016/j.isci.2024.108783] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), which has a prevalence of over 25% in adults, encompasses a wide spectrum of liver diseases. Metabolic-dysfunction associated steatotic liver disease (MASLD), the new term for NAFLD, is characterized by steatotic liver disease accompanied by cardiometabolic criteria, showing a strong correlation with metabolic diseases. Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disease affecting 4-21% of women of reproductive age. Numerous studies have indicated that NAFLD and PCOS often occur together. However, as MASLD is a new term, there is still a lack of reports describing the effects of MASLD on the development of PCOS. In this review article, we have summarized the complex and multifaceted connections between MASLD and PCOS. Understanding the pathogenesis and treatment methods could not only guide the clinical prevention, diagnosis, and treatment of PCOS in patients with MASLD, but also increase the clinical attention of reproductive doctors to MASLD.
Collapse
Affiliation(s)
- Qiuyu Xu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Metabolism and Regenerative Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Lu
- Institute of Metabolism and Regenerative Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Wu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Liu H, Tu M, Yin Z, Zhang D, Ma J, He F. Unraveling the complexity of polycystic ovary syndrome with animal models. J Genet Genomics 2024; 51:144-158. [PMID: 37777062 DOI: 10.1016/j.jgg.2023.09.012] [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: 07/03/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/02/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a highly familial and heritable endocrine disorder. Over half of the daughters born to women with PCOS may eventually develop their own PCOS-related symptoms. Progress in the treatment of PCOS is currently hindered by the complexity of its clinical manifestations and incomplete knowledge of its etiopathogenesis. Various animal models, including experimentally induced, naturally occurring, and spontaneously arising ones, have been established to emulate a wide range of phenotypical and pathological traits of human PCOS. These studies have led to a paradigm shift in understanding the genetic, developmental, and evolutionary origins of this disorder. Furthermore, emerging evidence suggests that animal models are useful in evaluating state-of-the-art drugs and treatments for PCOS. This review aims to provide a comprehensive summary of recent studies of PCOS in animal models, highlighting the power of these disease models in understanding the biology of PCOS and aiding high-throughput approaches.
Collapse
Affiliation(s)
- Huanju Liu
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Mixue Tu
- Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Zhiyong Yin
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Dan Zhang
- Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Zhejiang Provincial Clinical Research Center for Child Health, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Clinical Research Center on Birth Defect Prevention and Intervention of Zhejiang Province, Hangzhou, Zhejiang 310006, China.
| | - Jun Ma
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou, Zhejiang 310058, China.
| | - Feng He
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
13
|
Athar F, Karmani M, Templeman N. Metabolic hormones are integral regulators of female reproductive health and function. Biosci Rep 2024; 44:BSR20231916. [PMID: 38131197 PMCID: PMC10830447 DOI: 10.1042/bsr20231916] [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: 11/03/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023] Open
Abstract
The female reproductive system is strongly influenced by nutrition and energy balance. It is well known that food restriction or energy depletion can induce suppression of reproductive processes, while overnutrition is associated with reproductive dysfunction. However, the intricate mechanisms through which nutritional inputs and metabolic health are integrated into the coordination of reproduction are still being defined. In this review, we describe evidence for essential contributions by hormones that are responsive to food intake or fuel stores. Key metabolic hormones-including insulin, the incretins (glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1), growth hormone, ghrelin, leptin, and adiponectin-signal throughout the hypothalamic-pituitary-gonadal axis to support or suppress reproduction. We synthesize current knowledge on how these multifaceted hormones interact with the brain, pituitary, and ovaries to regulate functioning of the female reproductive system, incorporating in vitro and in vivo data from animal models and humans. Metabolic hormones are involved in orchestrating reproductive processes in healthy states, but some also play a significant role in the pathophysiology or treatment strategies of female reproductive disorders. Further understanding of the complex interrelationships between metabolic health and female reproductive function has important implications for improving women's health overall.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Muskan Karmani
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M. Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
14
|
Dar MA, Maqbool M, Ara I. The PCOS puzzle: putting the pieces together for optimal care. Int J Adolesc Med Health 2023; 35:299-311. [PMID: 37596861 DOI: 10.1515/ijamh-2023-0102] [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: 07/23/2023] [Accepted: 07/30/2023] [Indexed: 08/20/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a multifaceted hormonal disorder that has significant ramifications for both women's reproductive and metabolic well-being. This analysis aims to offer a thorough comprehension of PCOS by investigating the various contributing factors that are crucial for its effective management. We delve into the topic of hormonal imbalances, such as elevated androgens and disrupted estrogen-progesterone dynamics, and their effects on reproductive and metabolic health. Furthermore, we explore the intricate connection between insulin resistance, hyperinsulinemia, and PCOS, highlighting their pivotal role in metabolic dysfunction. Additionally, we examine fertility challenges, irregular menstrual patterns, and metabolic complications while also reviewing current treatment methodologies. Moreover, we address the latest research concerning genetic, environmental, and epigenetic influences on PCOS. By piecing together these essential elements, healthcare professionals can attain a comprehensive understanding of PCOS and deliver optimal care for those affected by the condition.
Collapse
Affiliation(s)
- Mohd Altaf Dar
- Department of Pharmacology, CT Institute of Pharmaceutical Sciences, PTU, Jalandhar, Punjab, India
| | - Mudasir Maqbool
- Department of Pharmaceutical Sciences, University Of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Irfat Ara
- Regional Research Institute of Unani Medicine, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
15
|
Rakic D, Joksimovic Jovic J, Jakovljevic V, Zivkovic V, Nikolic M, Sretenovic J, Nikolic M, Jovic N, Bicanin Ilic M, Arsenijevic P, Dimitrijevic A, Vulovic T, Ristic N, Bulatovic K, Bolevich S, Stijak L, Pantovic S. High Fat Diet Exaggerate Metabolic and Reproductive PCOS Features by Promoting Oxidative Stress: An Improved EV Model in Rats. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1104. [PMID: 37374308 DOI: 10.3390/medicina59061104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023]
Abstract
Background and Objectives: Polycystic ovary syndrome (PCOS) is a frequent multifactorial endocrinopathy affecting women in the reproductive period, often associated with infertility and metabolic disorders. The use of animal models helps to better understand etiopathogenesis, enabling the examination of the effects of certain drugs in order to discover the best possible therapeutic approach. We tried to investigate the additional effect of estradiol-valerate (EV) and high-fat diet (HFD) in female rats to explore PCOS-related alterations with special focus on oxidative stress. Materials and Methods: Animals were divided into three groups: control group (CTRL, n = 6), estradiol-valerate group (EV, n = 6), and estradiol-valerate group on HFD (EV + HFD, n = 6). PCOS was induced by single subcutaneous injection of long-acting EV in a dose of 4 mg/per rat. We tried to improve the metabolic characteristics of the PCOS animal model by adding HFD, so the CTRL and EV group had a regular diet, while the EV + HFD group had HFD during the induction period of 60 days. Results: We observed alterations of anthropometric parameters and hormonal disturbances, along with estrus cycle impairment reassembly to obese-type PCOS phenotype. Moreover, glucose metabolism was impaired after addition of HFD to EV protocol, contrary to EV administered alone. Histological analysis confirmed more numerous cystic follicles after the combination of EV and HFD protocol. The alterations of oxidative stress markers could be related to and serve as the mechanistic base for development of PCOS-related endocrine, reproductive, and metabolic properties. Conclusions: The additive effect of EV and HFD was obvious in the majority of the parameters observed. Our study strongly demonstrated metabolic as well as reproductive properties of PCOS in rats.
Collapse
Affiliation(s)
- Dejana Rakic
- Department of Gynecology and Obstetrics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- University Clinical Center Kragujevac, Zmaj Jovina 30, 34000 Kragujevac, Serbia
| | - Jovana Joksimovic Jovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University, 119146 Moscow, Russia
| | - Vladimir Zivkovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
- Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Maja Nikolic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
| | - Jasmina Sretenovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
| | - Marina Nikolic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
| | - Nikola Jovic
- Department of Gynecology and Obstetrics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- University Clinical Center Kragujevac, Zmaj Jovina 30, 34000 Kragujevac, Serbia
| | - Marija Bicanin Ilic
- Department of Gynecology and Obstetrics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- University Clinical Center Kragujevac, Zmaj Jovina 30, 34000 Kragujevac, Serbia
| | - Petar Arsenijevic
- Department of Gynecology and Obstetrics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- University Clinical Center Kragujevac, Zmaj Jovina 30, 34000 Kragujevac, Serbia
| | - Aleksandra Dimitrijevic
- Department of Gynecology and Obstetrics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- University Clinical Center Kragujevac, Zmaj Jovina 30, 34000 Kragujevac, Serbia
| | - Tatjana Vulovic
- University Clinical Center Kragujevac, Zmaj Jovina 30, 34000 Kragujevac, Serbia
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Natasa Ristic
- Department of Cytology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia
| | - Kristina Bulatovic
- Faculty of Medicine, University of Pristina in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia
| | - Sergej Bolevich
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University, 119146 Moscow, Russia
| | - Lazar Stijak
- Institute of Anatomy, School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Suzana Pantovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
16
|
Staszkiewicz R, Sobański D, Ulasavets U, Wieczorek J, Golec E, Marcol W, Grabarek BO. Evaluation of the concentration of selected elements in serum patients with intervertebral disc degeneration. J Trace Elem Med Biol 2023; 77:127145. [PMID: 36921371 DOI: 10.1016/j.jtemb.2023.127145] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 02/10/2023] [Accepted: 02/27/2023] [Indexed: 03/14/2023]
Abstract
Quantitative analysis of the trace element content of human intervertebral discs (IVDs) is essential because it can identify specific enzymes or metabolites that may be related to human intervertebral disc degeneration (IVDD). The goal of this study was to assess the concentrations of copper (Cu), iron (Fe), manganese (Mn), lead (Pb), zinc (Zn), sodium (Na), magnesium (Mg), potassium (K), phosphorus (P), and calcium (Ca) in serum samples obtained from patients with IVDD in comparison to healthy volunteers (a control group). The study group consisted of 113 Caucasian patients qualified by a specialist neurosurgeon for microdiscectomy. The control group consisted of 113 healthy volunteers who met the eligibility criteria for blood donors. The examined clinical material was the serum samples obtained from both groups.Based on the quantitative analysis of selected elements, there were statistically significantly (p 0.05) higher concentrations of Cu (1180 μg/L±800 μg/L vs. 1230 μg/L±750 μg/L), Zn (790 μg/L±300 μg/L vs. 850 μg/L±200 μg/L), and Mg (21730 μg/L±4360 μg/L vs. 23820 μg/L±4990 μg/L) in the serum of healthy volunteers compared to those in the study group. In addition, statistically significant changes were not detected in the concentrations of any elements among either sex in either the study or control group or in their body mass index (BMI) values (p > 0.05). In the serum samples from the study group, the strongest relationships were noted between the concentrations of Zn and Pb (r = 0.61), Zn and P (r = 0.69), Zn and Ca (r = 0.84), Zn and Cu (r = 0.83), Mg and Ca (r = 0.74), and Ca and P (r = 0.98).It has been indicated that, above all, the concentrations of Cu, Zn, Ca, and Mg depend on the advancement of radiological changes, according to the Pfirrmann scale. However, no influence on pain intensity was found, depending on the concentration of the assessed elements.The analysis indicates that the determination of serum Cu, Zn, Ca, and Mg concentrations may have diagnostic significance in predicting the onset of lumbosacral IVDD. The predictive evaluation of changes in the concentrations of selected elements in patients with degenerative lumbar IVD lesions appears to be a promising, cost-effective strategy.
Collapse
Affiliation(s)
- Rafał Staszkiewicz
- Department of Neurosurgery, 5th Military Clinical Hospital with the SP ZOZ Polyclinic in Krakow, 30-901 Krakow, Poland; Department of Histology, Cytophysiology and Embryology, Faculty of Medicine in Zabrze, Academy of Silesia in Katowice, 41-800 Zabrze, Poland.
| | - Dawid Sobański
- Department of Neurosurgery, St. Raphael Hospital, 30-693 Krakow, Poland; Department of Neurosurgery, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University, 30-705 Kraków, Poland
| | - Uladzislau Ulasavets
- Department of Neurosurgery, 5th Military Clinical Hospital with the SP ZOZ Polyclinic in Krakow, 30-901 Krakow, Poland
| | - Jerzy Wieczorek
- Department of Agricultural and Environmental Chemistry, University of Agriculture in Krakow, 31-120 Krakow, Poland
| | - Edward Golec
- Department of Trauma and Orthopedic Surgery, 5th Military Clinical Hospital, Kraków, Poland; Department of Rehabilitation in Orthopedics, Faculty of Motor Rehabilitation Bronisław Czech University of Physical Education in Kraków, Poland
| | - Wiesław Marcol
- Department of Physiology, School of Medicine in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; Department of Neurosurgery, Provincial Specialist Hospital No. 2 in Jastrzębie-Zdrój, 44-300 Jastrzębie-Zdrój, Poland
| | - Beniamin Oskar Grabarek
- Department of Neurosurgery, 5th Military Clinical Hospital with the SP ZOZ Polyclinic in Krakow, 30-901 Krakow, Poland; Department of Histology, Cytophysiology and Embryology, Faculty of Medicine in Zabrze, Academy of Silesia in Katowice, 41-800 Zabrze, Poland
| |
Collapse
|
17
|
Çıtar Dazıroğlu ME, Acar Tek N. The Effect on Inflammation of Adherence to the Mediterranean Diet in Polycystic Ovary Syndrome. Curr Nutr Rep 2023; 12:191-202. [PMID: 36719550 DOI: 10.1007/s13668-023-00451-6] [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] [Accepted: 09/30/2022] [Indexed: 02/01/2023]
Abstract
PURPOSE OF REVIEW Polycystic ovary syndrome (PCOS), which is common in women of reproductive age worldwide, is a syndrome that reduces the lifelong quality of life and poses a significant risk for various diseases. PCOS is a combination of symptoms of hyperandrogenism, oligo-anovulation, and polycystic ovarian morphology (PCOM). In PCOS, which is characterized by chronic low-grade inflammation, some inflammatory cytokines are increased. This review aimed to explain possible mechanisms of inflammation in PCOS and the effects of Mediterranean diet components on reducing this inflammation. RECENT FINDINGS Although the exact mechanisms of inflammation in PCOS are not yet fully known, it is stated that it is mediated by obesity, insulin resistance, and high androgen concentration. This inflammatory state negatively impacts the risk of future health problems and the quality of life of PCOS. Therefore, strategies to reduce inflammation are thought to be important. Dietary adjustments have important effects in reducing this inflammation and preventing disease. At this point, the Mediterranean diet, which has been proven to have a protective effect against many diseases, draws attention. Among the components of the Mediterranean diet, especially omega-3, antioxidants and dietary fiber may contribute to the reduction of inflammation through different mechanisms. PCOS is characterized by chronic low-grade inflammation, which increases women's risk of health problems, both now and in the future. Reducing inflammation is therefore extremely important, and it can be achieved with adherence to the Mediterranean diet. Inflammation pathways and the effect of the components of the Mediterranean diet in PCOS. AGE, advanced glycation end products; NF-κB, nuclear factor kappa-B. Obesity, insulin resistance, and hyperandrogenism may cause inflammation in PCOS through different mechanisms. Antioxidants, omega-3, and dietary fiber, which are the main components of the Mediterranean diet, may be effective in reducing this inflammation in PCOS. (Created with BioRender.com).
Collapse
Affiliation(s)
- Merve Esra Çıtar Dazıroğlu
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Emek, Ankara, Turkey.
| | - Nilüfer Acar Tek
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Emek, Ankara, Turkey
| |
Collapse
|
18
|
Mu L, Ye Z, Hu J, Zhang Y, Chen K, Sun H, Li R, Mao W, Long X, Zhang C, Lai Y, Liu J, Zhao Y, Qiao J. PPM1K-regulated impaired catabolism of branched-chain amino acids orchestrates polycystic ovary syndrome. EBioMedicine 2023; 89:104492. [PMID: 36863088 PMCID: PMC9986518 DOI: 10.1016/j.ebiom.2023.104492] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is one of the most common diseases with the coexistence of reproductive malfunction and metabolic disorders. Previous studies have found increased branched chain amino acid (BCAA) levels in women with PCOS. However, it remains unclear whether BCAA metabolism is causally associated with the risk of PCOS. METHODS The changes of BCAA levels in the plasma and follicular fluids of PCOS women were detected. Mendelian randomization (MR) approaches were used to explore the potential causal association between BCAA levels and the risk of PCOS. The function of the gene coding the protein phosphatase Mg2+/Mn2+-dependent 1K (PPM1K) was further explored by using Ppm1k-deficient mouse model and PPM1K down-regulated human ovarian granulosa cells. FINDINGS BCAA levels were significantly elevated in both plasma and follicular fluids of PCOS women. Based on MR, a potential direct, causal role for BCAA metabolism was revealed in the pathogenesis of PCOS, and PPM1K was detected as a vital driver. Ppm1k-deficient female mice had increased BCAA levels and exhibited PCOS-like traits, including hyperandrogenemia and abnormal follicle development. A reduction in dietary BCAA intake significantly improved the endocrine and ovarian dysfunction of Ppm1k-/- female mice. Knockdown of PPM1K promoted the conversion of glycolysis to pentose phosphate pathway and inhibited mitochondrial oxidative phosphorylation in human granulosa cells. INTERPRETATION Ppm1k deficiency-impaired BCAA catabolism causes the occurrence and development of PCOS. PPM1K suppression disturbed energy metabolism homeostasis in the follicular microenvironment, which provided an underlying mechanism of abnormal follicle development. FUNDING This study was supported by the National Key Research and Development Program of China (2021YFC2700402, 2019YFA0802503), the National Natural Science Foundation of China (81871139, 82001503, 92057107), the CAMS Innovation Fund for Medical Sciences (2019-I2M-5-001), Key Clinical Projects of Peking University Third Hospital (BYSY2022043), the China Postdoctoral Science Foundation (2021T140600), and the Collaborative Innovation Program of Shanghai Municipal Health Commission (2020CXJQ01).
Collapse
Affiliation(s)
- Liangshan Mu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China; Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenhong Ye
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Junhao Hu
- Transplantation Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yurong Zhang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Kai Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Haipeng Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Center for Cardiovascular Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Weian Mao
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyu Long
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Chunmei Zhang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yuchen Lai
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jun Liu
- Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China; Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China.
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China; Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinery Studies Peking University, Beijing 100871, China.
| |
Collapse
|
19
|
Multi-Nutrient Analysis of Dietary Macronutrients with All-Cause, Cardiovascular, and Cancer Mortality: Data from NHANES 1999-2014. Nutrients 2023; 15:nu15020345. [PMID: 36678215 PMCID: PMC9865351 DOI: 10.3390/nu15020345] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Macronutrients are a major component of the human diet. However, few studies have assessed their collective association with mortality. We sought to evaluate the associations of macronutrient intake with all-cause, cardiovascular, and cancer mortality in US adults using a multi-nutrient approach. This prospective cohort analysis used data from the National Health and Nutrition Examination Survey from the years 1999 to 2014. The participants included 33,681 US adults aged 20−85 years (52.5% female). The maximum follow-up time was 16.8 years, with a total of 4398 total deaths, including 772 cardiovascular deaths and 952 cancer deaths. The associations between mortality and dietary macronutrients were explored using three-dimensional generalized additive models, allowing for visual and statistical inference of complex nonlinear associations. Absolute macronutrient intake demonstrated a three-way interactive association with all-cause mortality (p < 0.001), cardiovascular mortality (p = 0.02), and cancer mortality (p = 0.05), adjusted for age, sex, ethnicity, socioeconomic status, dietary quality, and lifestyle. Compositionally, a high caloric diet composed of moderately high protein (20%), moderate fat (30%), and moderate carbohydrate (50%) levels was associated with the highest mortality risk. Across the total energy intake levels, lower mortality risk was observed in two separate regions consisting of higher protein (30%), higher carbohydrate (60%), and lower fat levels (10%) or lower protein (10%), moderate carbohydrate (45%), and higher fat levels (45%). These findings highlight a complex nonlinear and interactive association between macronutrients and all-cause mortality such that several distinct dietary compositions are associated with similarly high or low risk. Future research is needed to explore the drivers of these associations and whether they differ across varying dietary patterns and populations.
Collapse
|
20
|
Liu H, Li J, Chang X, He F, Ma J. Modeling Obesity-Associated Ovarian Dysfunction in Drosophila. Nutrients 2022; 14:nu14245365. [PMID: 36558524 PMCID: PMC9783805 DOI: 10.3390/nu14245365] [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: 11/21/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
We perform quantitative studies to investigate the effect of high-calorie diet on Drosophila oogenesis. We use the central composite design (CCD) method to obtain quadratic regression models of body fat and fertility as a function of the concentrations of protein and sucrose, two major macronutrients in Drosophila diet, and treatment duration. Our results reveal complex interactions between sucrose and protein in impacting body fat and fertility when they are considered as an integrated physiological response. We verify the utility of our quantitative modeling approach by experimentally confirming the physiological responses-including increased body fat, reduced fertility, and ovarian insulin insensitivity-expected of a treatment condition identified by our modeling method. Under this treatment condition, we uncover a Drosophila oogenesis phenotype that exhibits an accumulation of immature oocytes and a halt in the production of mature oocytes, a phenotype that bears resemblance to key aspects of the human condition of polycystic ovary syndrome (PCOS). Our analysis of the dynamic progression of different aspects of diet-induced pathophysiology also suggests an order of the onset timing for obesity, ovarian dysfunction, and insulin resistance. Thus, our study documents the utility of quantitative modeling approaches toward understanding the biology of Drosophila female reproduction, in relation to diet-induced obesity and type II diabetes, serving as a potential disease model for human ovarian dysfunction.
Collapse
Affiliation(s)
- Huanju Liu
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310058, China
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou 310058, China
| | - Jiajun Li
- ZJU-UOE Institute, Zhejiang University School of Medicine, Haining 314400, China
| | - Xinyue Chang
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310058, China
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou 310058, China
| | - Feng He
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310058, China
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou 310058, China
- Correspondence: (F.H.); (J.M.)
| | - Jun Ma
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310058, China
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou 310058, China
- Women’s Reproductive Health Research Laboratory of Zhejiang Province, Hangzhou 310006, China
- Zhejiang University-University of Toronto Joint Institute of Genetics and Genome Medicine, Hangzhou 310058, China
- Correspondence: (F.H.); (J.M.)
| |
Collapse
|
21
|
Lower Fiber Consumption in Women with Polycystic Ovary Syndrome: A Meta-Analysis of Observational Studies. Nutrients 2022; 14:nu14245285. [PMID: 36558444 PMCID: PMC9785338 DOI: 10.3390/nu14245285] [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: 10/24/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Polycystic ovary syndrome is a common endocrine disorder associated with metabolic abnormalities and gut microbiota dysbiosis. The deficiency of dietary fiber, a crucial nutrient in the daily diet, is also associated with a wide range of metabolic and reproductive abnormalities, as well as an altered gut microbial ecosystem. This study is a meta-analysis to summarize the available evidence on the dietary fiber intake level in PCOS patients. Databases of PubMed, Embase, Cochrane Library, Web of Science, and ClinicalTrials.gov were searched for observational studies, and 13 studies were finally included. The pooled standardized mean difference (SMD) with the 95% confidence interval (CI) of daily dietary fiber intake and total energy intake were calculated using the random-effects model. The pooled result (12 studies) on absolute dietary fiber intake showed that while there was no significant difference in the total energy intake [−0.17 (−0.44, 0.09), p = 0.208], the dietary fiber intake was significantly lower in PCOS women than those of controls [−0.32 (−0.50, −0.14), p < 0.001]. However, significant heterogeneity was detected across the studies (I2 = 65.6%, p = 0.001). Meta-regression suggested that geographic region and dietary assessment method may confer borderline significance of influence on the heterogeneity. The pooled result (two studies) on dietary fiber intake which adjusted for total energy intake, however, showed no significant difference [−2.11 (−4.77, 0.56), p = 0.122]. In subgroup analyses based on absolute dietary fiber intake, a lower dietary fiber intake in PCOS was observed in studies conducted in Asia, adopted food diary or records or food recall as the dietary assessment method, had a case−control study design, or used Rotterdam criteria for PCOS diagnosis. The difference in SMD was still significant in the adult subgroup or in studies matched or unmatched for age.
Collapse
|
22
|
Cai Z, He S, Liu R, Zhou L, Zhao L. Plumbagin rescues the granulosa cell's pyroptosis by reducing WTAP-mediated N6-methylation in polycystic ovary syndrome. J Ovarian Res 2022; 15:126. [PMID: 36463191 PMCID: PMC9719148 DOI: 10.1186/s13048-022-01058-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/02/2022] [Indexed: 12/05/2022] Open
Abstract
The survival of ovary granulosa cells (GC) is critical in the initiation and progression of polycystic ovary syndrome (PCOS) in females. Here, we found that the PCOS process is accompanied by massive GC pyroptosis resulting from Caspase-1 inflammasome activation. Administration of plumbagin, an effective compound isolated from plant medicine, can prevent the pyroptosis of GC and the onset of PCOS. Mechanistic study indicates the over-activation of the inflammasome in GC is due to the upregulation of WTAP, a key regulator of the RNA N6-methylase complex. WTAP mediates the mRNA N6-methylation of NLRP3 inflammasome component ASC and enhances ASC RNA stability, which results in the overactivation of the inflammasome in GCs from the PCOS model. Plumbagin treatment suppresses the WTAP-mediated N6-methylation of ASC mRNA and reduces the pyroptosis of GCs. This study supports the profound potential of plumbagin in PCOS treatment.
Collapse
Affiliation(s)
- Zhaowei Cai
- Reproductive Center, SSL central hospital of Dongguan, 1 Xianglong Road, Shilong, Dongguan, 523326 Guangdong China
| | - Shaojuan He
- grid.284723.80000 0000 8877 7471Department of clinical laboratory affiliated Dongguan hospital (Dongguan People’s Hospital), Southern Medical University, Dongguan, 523059 Guangdong China
| | - Rongju Liu
- Reproductive Center, SSL central hospital of Dongguan, 1 Xianglong Road, Shilong, Dongguan, 523326 Guangdong China
| | - Liling Zhou
- Reproductive Center, SSL central hospital of Dongguan, 1 Xianglong Road, Shilong, Dongguan, 523326 Guangdong China
| | - Li Zhao
- Reproductive Center, SSL central hospital of Dongguan, 1 Xianglong Road, Shilong, Dongguan, 523326 Guangdong China
| |
Collapse
|
23
|
Aflatounian A, Paris VR, Richani D, Edwards MC, Cochran BJ, Ledger WL, Gilchrist RB, Bertoldo MJ, Wu LE, Walters KA. Declining muscle NAD + in a hyperandrogenism PCOS mouse model: Possible role in metabolic dysregulation. Mol Metab 2022; 65:101583. [PMID: 36096453 PMCID: PMC9490589 DOI: 10.1016/j.molmet.2022.101583] [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: 10/26/2021] [Revised: 06/12/2022] [Accepted: 08/23/2022] [Indexed: 12/04/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder, defined by reproductive and endocrine abnormalities, with metabolic dysregulation including obesity, insulin resistance and hepatic steatosis. Recently, it was found that skeletal muscle insulin sensitivity could be improved in obese, post-menopausal, pre-diabetic women through treatment with nicotinamide mononucleotide (NMN), a precursor to the prominent redox cofactor nicotinamide adenine dinucleotide (NAD+). Given that PCOS patients have a similar endocrine profile to these patients, we hypothesised that declining NAD levels in muscle might play a role in the pathogenesis of the metabolic syndrome associated with PCOS, and that this could be normalized through NMN treatment. Here, we tested the impact of NMN treatment on the metabolic syndrome of the dihydrotestosterone (DHT) induced mouse model of PCOS. We observed lower NAD levels in the muscle of PCOS mice, which was normalized by NMN treatment. PCOS mice were hyperinsulinaemic, resulting in increased adiposity and hepatic lipid deposition. Strikingly, NMN treatment completely normalized these aspects of metabolic dysfunction. We propose that addressing the decline in skeletal muscle NAD levels associated with PCOS can normalize insulin sensitivity, preventing compensatory hyperinsulinaemia, which drives obesity and hepatic lipid deposition, though we cannot discount an impact of NMN on other tissues to mediate these effects. These findings support further investigation into NMN treatment as a new therapy for normalizing the aberrant metabolic features of PCOS.
Collapse
Affiliation(s)
- Ali Aflatounian
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Valentina Rodriguez Paris
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Dulama Richani
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Melissa C Edwards
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Blake J Cochran
- School of Medical Sciences, University of New South Wales Sydney, Sydney, NSW 2052, Australia
| | - William L Ledger
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Robert B Gilchrist
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Michael J Bertoldo
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia; School of Medical Sciences, University of New South Wales Sydney, Sydney, NSW 2052, Australia
| | - Lindsay E Wu
- School of Medical Sciences, University of New South Wales Sydney, Sydney, NSW 2052, Australia.
| | - Kirsty A Walters
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
24
|
Abstract
BACKGROUND The aim of this study was to find underlying genes and their interaction mechanism crucial to the polycystic ovarian syndrome (PCOS) by analyzing differentially expressed genes (DEGs) between PCOS and non-PCOS subjects. METHODS Gene expression data of PCOS and non-PCOS subjects were collected from gene expression omnibus (GEO) database. GEO2R were used to calculating P value and logFC. The screening threshold of DEGs was P < .05 and | FC | ≥ 1.2. GO annotation and Kyoto encyclopedia of genes and genomes (KEGG) signaling pathway enrichment analysis was performed by using DAVID (2021 Update). The protein-protein interaction (PPI) network of DEGs was constructed by using the STRING database, and the hub genes were recognized through Hubba plugin of Cytoscape software. RESULTS PCOS and non-PCOS subjects shared a total of 174 DGEs, including 14 upregulated and 160 downregulated genes. The GO biological processes enriched by DEGs mainly involved actin cytoskeleton organization, positive regulation of NF-κB signaling pathway, and positive regulation of canonical Wnt signaling pathway. The DEGs were significantly enriched in cytoplasm, nucleus and cytosol. Their molecular functions mainly focused on protein binding, calmodulin binding and glycerol-3-phosphate dehydrogenase activity. The PI3K/Akt signaling pathway and glycosaminoglycan biosynthesis were highlighted as critical pathways enriched by DEGs. 10 hub genes were screened from the constructed PPI network, of which EGF, FN1 and TLR4 were mainly enriched in the PI3K/Akt signaling pathway. CONCLUSION In this study, a total of 174 DEGs and 10 hub genes were identified as new candidate targets for insulin resistance (IR) in PCOS individuals, which may provide a new direction for developing novel treatment strategies for PCOS.
Collapse
Affiliation(s)
- Fei Zhou
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yuling Xing
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Tiantian Cheng
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Linlin Yang
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Huijuan Ma
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei, China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
- *Correspondence: Huijuan Ma, Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang 050017, Hebei, China (e-mail: )
| |
Collapse
|
25
|
Zhang J, Zhang H, Xin X, Zhu Y, Ye Y, Li D. Efficacy of Flavonoids on Animal Models of Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis. Nutrients 2022; 14:nu14194128. [PMID: 36235780 PMCID: PMC9571610 DOI: 10.3390/nu14194128] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/23/2022] [Accepted: 09/30/2022] [Indexed: 12/09/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common gynecological endocrinopathies. Evidence suggest that flavonoids have beneficial effects on endocrine and metabolic diseases, including PCOS. However, high-quality clinical trials are lacking. We aimed to conduct a systematic review and meta-analysis of experimental studies to determine the flavonoids' effects in animal models of PCOS. Three electronic databases including PubMed, Scopus, and Web of Science were systematically searched from their inception to March 2022. The Systematic Review Center for Laboratory Animal Experimentation's risk of bias tool was used to assess methodological quality. The standardized mean difference was calculated with 95% confidence intervals as the overall effects. R was used for all statistical analyses. This study was registered in PROSPERO (registration number: CRD42022328355). A total of eighteen studies, including 300 animals, met the inclusion criteria. Our analyses demonstrated that, compared to control groups, flavonoid groups showed a significantly lower count of atretic follicles and cystic follicles and the count of corpus luteum was higher. A significant reduction in the luteinizing hormone (LH), LH/follicle-stimulating hormone (FSH), and free testosterone were observed in intervention groups. Nevertheless, there was no significant difference in the effects of flavonoids on the level of FSH, estradiol, and progesterone. Subgroup analyses indicated that the type of flavonoid, dose, duration of administration, and PCOS induction drug were relevant factors that influenced the effects of intervention. Current evidence supports the positive properties of flavonoids on ovarian histomorphology and hormonal status in animal models of PCOS. These data call for more randomized controlled trials and further experimental studies investigating the mechanism in more depth.
Collapse
Affiliation(s)
| | | | | | | | - Yang Ye
- Correspondence: (Y.Y.); (D.L.)
| | - Dong Li
- Correspondence: (Y.Y.); (D.L.)
| |
Collapse
|
26
|
Wu YX, Yang XY, Han BS, Hu YY, An T, Lv BH, Lian J, Wang TY, Bao XL, Gao L, Jiang GJ. Naringenin regulates gut microbiota and SIRT1/ PGC-1ɑ signaling pathway in rats with letrozole-induced polycystic ovary syndrome. Biomed Pharmacother 2022; 153:113286. [PMID: 35724506 DOI: 10.1016/j.biopha.2022.113286] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/31/2022] [Accepted: 06/09/2022] [Indexed: 11/02/2022] Open
Abstract
PURPOSE To evaluate the effect of naringenin on improving PCOS and explore the mechanism. METHODS Firstly, we carried out differential gene expression analysis from transcriptome sequencing data of human oocyte to screen the KEGG pathway, then the PCOS-like rat model was induced by letrozole. They were randomly divided into four groups: Normal group (N), PCOS group (P), Diane-35 group (D), and Naringenin group (Nar). The changes of estrus cycle, body weight, ovarian function, serum hormone levels, glucose metabolism, along with the expression of SIRT1, PGC-1ɑ, claudin-1 and occludin of the ovary and colon were investigated. Furthermore, the composition of the gut microbiome of fecal was tested. RESULTS By searching the KEGG pathway in target genes, we found that at least 15 KEGG pathways are significantly enriched in the ovarian function, such as AMPK signaling pathway, insulin secretion, and ovarian steroidogenesis. Interestingly, naringenin supplementation significantly reduced body weight, ameliorated hormone levels, improved insulin resistance, and mitigated pathological changes in ovarian tissue, up-regulated the expression of PGC-1ɑ, SIRT1, occludin and claudin-1 in colon. In addition, we also found that the abundance of Prevotella and Gemella was down-regulated, while the abundance of Butyricimonas, Lachnospira, Parabacteroides, Butyricicoccus, Streptococcus, Coprococcus was up-regulated. CONCLUSION Our data suggest that naringenin exerts a treatment PCOS effect, which may be related to the modulation of the gut microbiota and SIRT1/PGC-1ɑ signaling pathway. Our research may provide a new perspective for the treatment of PCOS and related diseases.
Collapse
Affiliation(s)
- Yan-Xiang Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiu-Yan Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bao-Sheng Han
- Maternity and Child Care Hospital of North China University of Science and Technology, Tangshan, China
| | - Yuan-Yuan Hu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tian An
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bo-Han Lv
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Juan Lian
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ting-Ye Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xue-Li Bao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Lin Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Guang-Jian Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
27
|
Xiong T, Rodriguez Paris V, Edwards MC, Hu Y, Cochran BJ, Rye KA, Ledger WL, Padmanabhan V, Handelsman DJ, Gilchrist RB, Walters KA. Androgen signaling in adipose tissue, but less likely skeletal muscle, mediates development of metabolic traits in a PCOS mouse model. Am J Physiol Endocrinol Metab 2022; 323:E145-E158. [PMID: 35658542 DOI: 10.1152/ajpendo.00418.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common, multifactorial disorder characterized by endocrine, reproductive, and metabolic dysfunction. As the etiology of PCOS is unknown, there is no cure and symptom-oriented treatments are suboptimal. Hyperandrogenism is a key diagnostic trait, and evidence suggests that androgen receptor (AR)-mediated actions are critical to PCOS pathogenesis. However, the key AR target sites involved remain to be fully defined. Adipocyte and muscle dysfunction are proposed as important sites involved in the manifestation of PCOS traits. We investigated the role of AR signaling in white adipose tissue (WAT), brown adipose tissue (BAT), and skeletal muscle in the development of PCOS in a hyperandrogenic PCOS mouse model. As expected, dihydrotestosterone (DHT) exposure induced key reproductive and metabolic PCOS traits in wild-type (WT) females. Transplantation of AR-insensitive (AR-/-) WAT or BAT from AR knockout females (ARKO) into DHT-treated WT mice ameliorated some metabolic PCOS features, including increased body weight, adiposity, and adipocyte hypertrophy, but not reproductive PCOS traits. In contrast, DHT-treated ARKO female mice transplanted with AR-responsive (AR+/+) WAT or BAT continued to resist developing PCOS traits. DHT-treated skeletal muscle-specific AR knockout females (SkMARKO) displayed a comparable phenotype with that of DHT-treated WT females, with full development of PCOS traits. Taken together, these findings infer that both WAT and BAT, but less likely skeletal muscle, are key sites of AR-mediated actions involved in the experimental pathogenesis of metabolic PCOS traits. These data further support targeting adipocyte AR-driven pathways in future research aimed at developing novel therapeutic interventions for PCOS.NEW & NOTEWORTHY Hyperandrogenism is a key feature in the pathogenesis of polycystic ovary syndrome (PCOS); however, the tissue sites of androgen receptor (AR) signaling are unclear. In this study, AR signaling in white and brown adipose tissue, but less likely in skeletal muscle, was found to be involved in the development of metabolic PCOS traits, highlighting the importance of androgen actions in adipose tissue and obesity in the manifestation of metabolic disturbances.
Collapse
Affiliation(s)
- Ting Xiong
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Valentina Rodriguez Paris
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Melissa C Edwards
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Ying Hu
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Blake J Cochran
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - William L Ledger
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | | | - David J Handelsman
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Robert B Gilchrist
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Kirsty A Walters
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
28
|
Rodriguez Paris V, Wong XYD, Solon-Biet SM, Edwards MC, Aflatounian A, Gilchrist RB, Simpson SJ, Handelsman DJ, Kaakoush NO, Walters KA. The interplay between PCOS pathology and diet on gut microbiota in a mouse model. Gut Microbes 2022; 14:2085961. [PMID: 35787106 PMCID: PMC9450977 DOI: 10.1080/19490976.2022.2085961] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The gut microbiome has been implicated in polycystic ovary syndrome (PCOS) pathophysiology. PCOS is a disorder with reproductive, endocrine and metabolic irregularities, and several studies report that PCOS is associated with a decrease in microbial diversity and composition. Diet is an important regulator of the gut microbiome, as alterations in macronutrient composition impact the balance of gut microbial communities. This study investigated the interplay between macronutrient balance and PCOS on the gut microbiome of control and dihydrotestosterone (DHT)-induced PCOS-like mice exposed to diets that varied in protein (P), carbohydrate (C) and fat (F) content. The amount of dietary P, C and F consumed significantly altered alpha (α) and beta (β) diversity of the gut microbiota of control and PCOS-like mice. However, α-diversity between control and PCOS-like mice on the same diet did not differ significantly. In contrast, β-diversity was significantly altered by PCOS pathology. Further analysis identified an operational taxonomic unit (OTU) within Bacteroides (OTU3) with 99.2% similarity to Bacteroides acidifaciens, which is inversely associated with obesity, to be significantly decreased in PCOS-like mice. Additionally, this study investigated the role of the gut microbiome in the development of PCOS traits, whereby PCOS-like mice were transplanted with healthy fecal microbiota from control mice. Although the PCOS gut microbiome shifted toward that of control mice, PCOS traits were not ameliorated. Overall, these findings demonstrate that while diet exerts a stronger influence over gut microbiota diversity than PCOS pathology, overall gut microbiota composition is affected by PCOS pathology.
Collapse
Affiliation(s)
- Valentina Rodriguez Paris
- Fertility & Research Centre, School of Clinical Medicine, University of New South Wales Sydney, Sydney, NSW, Australia,CONTACT Valentina Rodriguez Paris Fertility & Research Centre, School of Women’s and Children’s Health, University of New South Wales Sydney, NSW2052, Australia
| | - Xin Yi Denise Wong
- Fertility & Research Centre, School of Clinical Medicine, University of New South Wales Sydney, Sydney, NSW, Australia
| | | | - Melissa C Edwards
- Fertility & Research Centre, School of Clinical Medicine, University of New South Wales Sydney, Sydney, NSW, Australia,ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Ali Aflatounian
- Fertility & Research Centre, School of Clinical Medicine, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Robert B Gilchrist
- Fertility & Research Centre, School of Clinical Medicine, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | | | - Nadeem O Kaakoush
- School of Medical Sciences, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Kirsty A Walters
- Fertility & Research Centre, School of Clinical Medicine, University of New South Wales Sydney, Sydney, NSW, Australia,ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
29
|
Zheng Y, He J, Yang D, Dai F, Yuan M, Liu S, Jia Y, Cheng Y. Irisin reduces the abnormal reproductive and metabolic phenotypes of PCOS by regulating the activity of brown adipose tissue in mice. Biol Reprod 2022; 107:1046-1058. [PMID: 35713297 PMCID: PMC9562123 DOI: 10.1093/biolre/ioac125] [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] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/05/2022] [Accepted: 06/08/2022] [Indexed: 11/14/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disease in women, with clinical manifestations of anovulation and hyperandrogenaemia. The treatment of PCOS mainly focuses on improving clinical symptoms, such as insulin sensitivity or menstrual disorder, through drug treatment. However, due to the pathogenesis diversity of PCOS, there is still a lack of effective treatment in clinics. Metabolic disorder is the key factor in the occurrence of PCOS. Brown adipose tissue (BAT) is a special adipose tissue in the human body that can participate in metabolic balance by improving heat production. BAT has been demonstrated to be an important substance involved in the metabolic disorder of PCOS. Although increasing evidence indicates that BAT transplantation can improve the symptoms of PCOS, it is difficult to achieve BAT transplantation at present due to technical limitations. Stimulation of BAT activation by exogenous substances may be an effective alternative therapy for PCOS. In this study, we investigated the effects of Irisin on dehydroepiandrosterone (DHEA)-induced PCOS in mice and evaluated the effect of Irisin on serum hormone levels and changes in body temperature, body weight and ovarian morphology. In our study, we found that Irisin can enhance the thermogenesis and insulin sensitivity of PCOS mice by activating the function of BAT. In addition, Irisin treatment can correct the menstrual cycle of PCOS mice, improve the serum steroid hormone disorder status, and reduce the formation of ovarian cystic follicles. In conclusion, our results showed that Irisin treatment significantly improved the metabolic disorder of PCOS and may provide a new and alternative therapy for the treatment of this pathology.
Collapse
Affiliation(s)
- Yajing Zheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Juan He
- Department of Obstetrics and Gynecology Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Mengqin Yuan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shiyi Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yifan Jia
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
30
|
Zia A, Hakim S, Khan AU, Bey A, Ateeq H, Parveen S, Khalid S, Yusufi F. Bone markers and bone mineral density associates with periodontitis in females with poly-cystic ovarian syndrome. J Bone Miner Metab 2022; 40:487-497. [PMID: 35072780 DOI: 10.1007/s00774-021-01302-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/10/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Studies suggest an association between poly-cystic ovarian syndrome (PCOS) and chronic periodontitis (CP), both being inflammatory conditions. However, insufficient evidence assesses the impact of this inflammation on bone metabolism and bone turnover markers (BTMs). The present study aimed to determine the association between BTMs, bone mineral density (BMD), and clinical periodontal parameters in PCOS women with CP. MATERIALS AND METHODS Three groups, each with 40 newly diagnosed (1) PCOS+CP, (2) PCOS alone, (3) CP alone, and fourth group (n = 20) systemically and periodontally healthy females aged 18-30 years were included in the study. Full mouth clinical periodontal parameters, C-terminal telopeptides of type I collagen (CTX), bone alkaline phosphatase (ALP), BMD and 25-hydroxyvitamin D (VD) were recorded for all. RESULTS Low BMD (0.89 ± 0.11 g/cm2), increased CTX levels (2.76 ± 4.64 ng/ml), decreased bone ALP levels (11.09 ± 6.86 ng/ml), higher VD levels (289.02 ± 168.28 nmol/l) and poor clinical periodontal status were observed in PCOS + CP females. BMD-spine showed weak positive correlation with CTX, bone ALP, VD (r = 0.02, r = 0.07, r = 0.15, respectively) in PCOS + CP group. ANCOVA depicted covariates had no confounding effect. Multiple regression model explained 21.0% for BMD-spine and 12.7% for BMD-femur of total variability signifying association with all measured parameters among all groups. CONCLUSION Enhanced inflammatory thrust by periodontitis increases CTX levels and decreases bone ALP and BMD levels in women with PCOS. Screening PCOS women for periodontal disease and vice versa may have a direct bearing on overall bone health.
Collapse
Affiliation(s)
- Afaf Zia
- Department of Periodontics and Community Dentistry, Dr. Ziauddin Ahmed Dental College (DRZADC), Aligarh Muslim University (AMU), Aligarh, 202002, Uttar Pradesh, India.
| | - S Hakim
- Department of Obstetrics and Gynaecology, Jawaharlal Nehru Medical College and Hospital (JNMCH), AMU, Aligarh, Uttar Pradesh, India
| | - A U Khan
- Interdisciplinary Biotechnology Unit, AMU, Aligarh, Uttar Pradesh, India
| | - A Bey
- Department of Periodontics and Community Dentistry, Dr. Ziauddin Ahmed Dental College (DRZADC), Aligarh Muslim University (AMU), Aligarh, 202002, Uttar Pradesh, India
| | - H Ateeq
- Department of Biochemistry, AMU, Aligarh, Uttar Pradesh, India
| | - S Parveen
- Department of Obstetrics and Gynaecology, Jawaharlal Nehru Medical College and Hospital (JNMCH), AMU, Aligarh, Uttar Pradesh, India
| | - S Khalid
- Interdisciplinary Biotechnology Unit, AMU, Aligarh, Uttar Pradesh, India
| | - Fnk Yusufi
- Department of Statistics and Operations, AMU, Aligarh, Uttar Pradesh, India
| |
Collapse
|
31
|
Na Z, Jiang H, Meng Y, Song J, Feng D, Fang Y, Shi B, Li D. Association of galactose and insulin resistance in polycystic ovary syndrome: A case-control study. EClinicalMedicine 2022; 47:101379. [PMID: 35480079 PMCID: PMC9035629 DOI: 10.1016/j.eclinm.2022.101379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is closely linked to metabolic disorders. Recent reports have identified galactose as having strong associations with metabolic disorders, however, the correlation between galactose and PCOS remains largely unknown. METHODS The serum galactose levels of 104 patients with PCOS and 98 controls were measured, and their relationships with several metabolic parameters were analyzed. The study took place at the Center for Reproductive Medicine at Shengjing Hospital of China Medical University, Shenyang, China from July 13 to Oct 20, 2020. The relationships between serum galactose and PCOS as well as PCOS-related insulin resistance were investigated via logistic regression analyses, and the performance of serum galactose as a potential biomarker for PCOS was evaluated using receiver operating characteristic curve analysis. FINDINGS Higher serum galactose levels were observed in the patients with PCOS than in the controls (p = 0.001). There was still a correlation between circulating galactose levels and PCOS after adjusting for covariates (p = 0.002; odds ratio (OR), 1.133; 95% confidence interval (CI) 1.047-1.227). Serum galactose levels were shown to be most closely related to the fasting serum insulin level (r = 0.370, p = 0.001) and were higher in the insulin-resistant subgroup than in the non-insulin-resistant subgroup of patients with PCOS (p = 0.001). There was no difference in serum galactose levels between the insulin-resistant and non-insulin-resistant subgroups of women in the control group (p > 0.05). Furthermore, higher serum galactose levels were shown to be associated with insulin resistance in PCOS (p = 0.004; OR, 26.017; 95% CI, 2.907-232.810). The area under the curve for galactose-mediated prediction of PCOS was 80.0%, with a sensitivity of 71.0% and a specificity of 86.4%. INTERPRETATION Higher circulating galactose levels correlate with PCOS and PCOS-related insulin resistance; therefore, it may serve as a potential biomarker for patients with PCOS. These findings require further validation in a study with a larger sample size. FUNDING National Natural Science Foundation of China (No. 82,071,607 and 32,100,691); LiaoNing Revitalization Talents Program (No. XLYC1907071); Fok Ying Tung Education Foundation (No. 151,039); Key Research and Development Program of Liaoning Province (NO. 2,018,225,062); Outstanding Scientific Fund of Shengjing Hospital (No. 202,003).
Collapse
Affiliation(s)
- Zhijing Na
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, No.36, SanHao Street, Shenyang 110004, China
| | - Hongyu Jiang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, No.36, SanHao Street, Shenyang 110004, China
| | - Yaxin Meng
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, No.36, SanHao Street, Shenyang 110004, China
| | - Jiahui Song
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, No.36, SanHao Street, Shenyang 110004, China
| | - Di Feng
- Education Center for Clinical Skill Practice, China Medical University, Shenyang 110122, China
| | - Yuanyuan Fang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, No.36, SanHao Street, Shenyang 110004, China
| | - Bei Shi
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, No.36, SanHao Street, Shenyang 110004, China
- Department of Physiology, School of Life Sciences, China Medical University, Shenyang 110122, China
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, No.36, SanHao Street, Shenyang 110004, China
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang 110122, China
| |
Collapse
|
32
|
Han Y, Lin B, Lu W, Wang X, Tang W, Tao X, Cai H, He C, Liu C. Time-restricted feeding improves metabolic and endocrine profiles in mice with polycystic ovary syndrome. Front Endocrinol (Lausanne) 2022; 13:1057376. [PMID: 36619541 PMCID: PMC9815607 DOI: 10.3389/fendo.2022.1057376] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVES Polycystic ovary syndrome (PCOS) is one of the most common endocrinopathy disorders in premenopausal women, which is characterized by hyperandrogenemia, anovulation, and polycystic ovarian morphology (PCOM). Time-restricted feeding (TRF) is a new intermittent restriction dietary pattern, which has been shown to have positive benefits on obesity and glycolipid metabolism disorders. We aimed to explore the effect of the feeding regimen (ad libitum vs. TRF) on the glycolipid metabolism and reproductive endocrine disorders in a PCOS mouse model. METHODS PCOS mouse model was induced by continuous subcutaneous administration of dihydrotestosterone for 21 days. Mice were fed a high-fat diet (HFD) for 8 weeks on an ad libitum or time- restricted diet (from 10:30 p.m. to 6:30 a.m.). RESULTS Compared to control mice, PCOS mice that received TRF treatment had significantly lower body weight, reduced adiposity, lower area under the curve (AUC) of glucose response in the oral glucose tolerance test (OGTT), and lower AUC in the insulin tolerance test (ITT). TRF also ameliorated lipid metabolism, as shown by a reduction in plasma lipid profiles (triglycerides and cholesterol) and the triglyceride content in the liver of PCOS mice. In terms of reproduction, the plasma androgen level, plasma estrogen (E2) level, and luteinizing hormone (LH)/follicle stimulating hormone (FSH) ratio in PCOS mice were significantly reduced after 8 weeks of TRF treatment. In addition, ovarian histology showed that TRF inhibits cyst formation and promotes corpus luteum formation. CONCLUSION In conclusion, TRF improved metabolic and endocrine profiles in mice with PCOS.
Collapse
Affiliation(s)
- Yan Han
- School of Medicine, Xiamen University, Xiamen, China
| | - Baiwei Lin
- School of Medicine, Xiamen University, Xiamen, China
| | - Wenjing Lu
- School of Medicine, Xiamen University, Xiamen, China
| | - Xu Wang
- School of Life Science, Anhui Medical University, Hefei, China
- Shanghai Key Laboratory of Metabolic Remodeling and Disease, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenshuai Tang
- Shanghai Key Laboratory of Metabolic Remodeling and Disease, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinge Tao
- School of Medicine, Xiamen University, Xiamen, China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Chunmei He
- Department of Endocrinology and Diabetes, the First Affiliated Hospital of Xiamen University, School of medicine, Xiamen University, Xiamen, China
- *Correspondence: Changqin Liu, ; Chunmei He,
| | - Changqin Liu
- Department of Endocrinology and Diabetes, the First Affiliated Hospital of Xiamen University, School of medicine, Xiamen University, Xiamen, China
- Fujian Province Key Laboratory of Diabetes Translational Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
- *Correspondence: Changqin Liu, ; Chunmei He,
| |
Collapse
|
33
|
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: 45] [Impact Index Per Article: 15.0] [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
|
34
|
Chen W, Pang Y. Metabolic Syndrome and PCOS: Pathogenesis and the Role of Metabolites. Metabolites 2021; 11:metabo11120869. [PMID: 34940628 PMCID: PMC8709086 DOI: 10.3390/metabo11120869] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine diseases among women of reproductive age and is associated with many metabolic manifestations, such as obesity, insulin resistance (IR) and hyperandrogenism. The underlying pathogenesis of these metabolic symptoms has not yet been fully elucidated. With the application of metabolomics techniques, a variety of metabolite changes have been observed in the serum and follicular fluid (FF) of PCOS patients and animal models. Changes in metabolites result from the daily diet and occur during uncommon physiological routines. However, some of these metabolite changes may provide evidence to explain possible mechanisms and new approaches for prevention and therapy. This article reviews the pathogenesis of PCOS metabolic symptoms and the relationship between metabolites and the pathophysiology of PCOS. Furthermore, the potential clinical application of some specific metabolites will be discussed.
Collapse
Affiliation(s)
- Weixuan Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China;
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing 100191, China
| | - Yanli Pang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China;
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing 100191, China
- Correspondence:
| |
Collapse
|
35
|
Shang Y, Zhou H, He R, Lu W. Dietary Modification for Reproductive Health in Women With Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne) 2021; 12:735954. [PMID: 34790167 PMCID: PMC8591222 DOI: 10.3389/fendo.2021.735954] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Objective Diet has been reported as the first-line management of polycystic ovary syndrome (PCOS). However, the relationship between diet and fertility in PCOS is still controversial. This meta-analysis aimed to evaluate whether diet could promote reproductive health in women with PCOS while providing evidence-based nutrition advice for clinical practice. Methods Seven databases, including Cochrane Central Register of Controlled Trials, PubMed, Embase, Web of Science, and some Chinese database, were searched up to January 31, 2021. Randomized controlled trials evaluating the effects of diet in women with PCOS were included. Based on a preregistered protocol (PROSPERO CRD42019140454), the systematic review was performed following the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. Two reviewers made study selection, data extraction and bias assessment independently. Risk ratios and mean difference with 95% confidence intervals were assessed by a random-effects model. Statistical heterogeneity within comparisons was evaluated by Cochran's Q test and quantified by the I-squared (I2 ) statistic. Results Twenty RCTs with 1113 participants were included. Results showed diet significantly related to improved fertility outcomes (increasing clinical pregnancy, ovulation and menstrual regularity rate; reducing miscarriage rate), reproductive endocrine [increasing sex hormone-binding globulin (SHBG); decreasing Anti-Müllerian Hormone (AMH), free androgen index (FAI), total testosterone (T)] and clinical hyperandrogenism (hirsutism assessed by Ferriman-Gallwey score) in PCOS. Specifically, subgroup analyses indicated low-carbohydrate diets were superior in optimizing reproductive outcomes and calorie restriction was critical in ameliorating hyperandrogenism. Additionally, the positive effects were associated with the treatment duration. The longer the duration, the greater the improvement was. Conclusion Overall, diet is an effective intervention for improving fertility health, thus professional and dynamic dietary advice should be offered to all PCOS patients, based on the changeable circumstances, personal needs and expectations of the individuals.
Collapse
Affiliation(s)
- Yujie Shang
- Department of Gynecology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huifang Zhou
- Department of Gynecology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ruohan He
- Maternal and Child Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wentian Lu
- Department of Gynecology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
36
|
The protective effect of sulforaphane against oxidative stress in granulosa cells of patients with polycystic ovary syndrome (PCOS) through activation of AMPK/AKT/NRF2 signaling pathway. Reprod Biol 2021; 21:100563. [PMID: 34678578 DOI: 10.1016/j.repbio.2021.100563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/22/2021] [Accepted: 09/16/2021] [Indexed: 01/04/2023]
Abstract
Increased production of reactive oxygen species (ROS) in granulosa cells (GCs) causes oxidative stress (OS) and plays a role in pathogenesis of polycystic ovary syndrome (PCOS). Sulforaphane (SFN) has received a great deal of attention as potent antioxidant because of its ability to induce expression of antioxidant enzymes through nuclear factor (erythroid-derived 2)-like 2 (NRF2) signaling pathway. Therefore, the present study was done to investigate the protective effect of SFN against OS in granulosa-lutein cells (GLCs) of patients with PCOS through activation of AMP-activated protein kinase (AMPK)/AKT/NRF2 signaling pathway. GLCs were isolated from patients with PCOS and healthy fertile women, as control group, during egg retrieval procedure. Level of intracellular ROS and apoptosis was determined in the isolated cells. For investigating the protective effect of SFN against ROS production and apoptosis in GLCs, the cells were cultured for 24 h in the presence or absence of SFN. Finally, expression of AMPK, AKT, and NRF2 proteins and genes was evaluated by western blotting and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. The results indicated the increased ROS and apoptosis levels in GLCs isolated from patients with PCOS compared to the control group. Addition of SFN to culture medium of GLCs of patients with PCOS reduced intracellular ROS and apoptosis levels, and increased expression of AMPK, AKT, and NRF2 proteins and genes. Our findings demonstrated the protective effect of SFN against OS by lowering level of ROS and apoptosis possibly through activation of AMPK, AKT, and NRF2 proteins and genes expression.
Collapse
|
37
|
Barrea L, Frias-Toral E, Verde L, Ceriani F, Cucalón G, Garcia-Velasquez E, Moretti D, Savastano S, Colao A, Muscogiuri G. PCOS and nutritional approaches: Differences between lean and obese phenotype. Metabol Open 2021; 12:100123. [PMID: 34622189 PMCID: PMC8479825 DOI: 10.1016/j.metop.2021.100123] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 01/15/2023] Open
Abstract
Polycystic Ovary Syndrome (PCOS) is an endocrine/metabolic disorder with an ever-increasing prevalence. It has various clinical characteristics; the cardinals are androgen excess, oligo-anovulatory infertility, polycystic ovaries, insulin resistance (IR), and cardiometabolic alterations. These disturbances are a consequence of PCOS's complex etiology. PCOS is mainly related to women with obesity; however, there are many PCOS lean patients too. Even though they share some aspects in their metabolic profiles, each group has individual differences in body composition and other parameters. Thus, in order to achieve successful therapeutic strategies, they should be tailored to these details. The authors reviewed PubMed's updated and related publications about body composition and nutritional strategies for PCOS lean and obese patients. As previous reports have determined, dietary patterns are essential in PCOS treatment. Several diets have been studied to control and improve IR, infertility, and cardiometabolic dysfunctions in PCOS. This review will explain the specific features in metabolic characterization and body composition among these patients. Finally, the diverse nutritional strategies used in women with PCOS will be analyzed depending on their lean or obese phenotype.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento di Scienze Umanistiche, Università Telematica Pegaso, 80143, Napoli, Italy.,Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", Federico II, Napoli, Italy
| | - Evelyn Frias-Toral
- Universidad Católica Santiago de Guayaquil, Av. Pdte. Carlos Julio Arosemena Tola, Guayaquil, 090615, Ecuador
| | - Ludovica Verde
- Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, Italy
| | - Florencia Ceriani
- Escuela de Nutrición, Universidad de la República, Montevideo, Uruguay
| | - Gabriela Cucalón
- ESPOL Polytechnic University, Escuela Superior Politécnica del Litoral, ESPOL, Lifescience Faculty, Campus Gustavo Galindo Km. 30.5 Vía Perimetral, P.O. Box 09-01-5863, Guayaquil, Ecuador
| | | | - Dino Moretti
- Hospital "Eva Perón", Avenida San Martín, 1645, G. Baigorria, Santa Fe, Argentina
| | - Silvia Savastano
- Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, Italy
| | - Annamaria Colao
- Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", Federico II, Napoli, Italy.,Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, Italy
| | - Giovanna Muscogiuri
- Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", Federico II, Napoli, Italy.,Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, Italy
| |
Collapse
|
38
|
Zhu Y, Qiu L, Jiang F, Găman MA, Abudoraehem OS, Okunade KS, Zhang M. The effect of dehydroepiandrosterone (DHEA) supplementation on estradiol levels in women: A dose-response and meta-analysis of randomized clinical trials. Steroids 2021; 173:108889. [PMID: 34246664 DOI: 10.1016/j.steroids.2021.108889] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/27/2021] [Accepted: 07/05/2021] [Indexed: 12/17/2022]
Abstract
Estradiol, an estrogen steroid hormone, serves as the dominant female hormone and its levels fluctuate during lifetime. In women, after the menopause, all estrogens and almost all androgens are locally developed in the peripheral tissues from dehydroepiandrosterone (DHEA). However, the effect of DHEA supplementation on estradiol levels in women is unclear as previously published data has resulted in conflicting findings. Thus, we conducted the present dose-response meta-analysis of randomized controlled trials (RCTs) evaluating the influence of DHEA on estradiol concentrations in women. The PubMed/Medline, Embase, Web of Science and Scopus databases were systematically searched for articles published on this topic until May 10, 2021. No time or language restrictions were applied. The data were expressed as weighted mean differences (WMDs) and 95% confidence intervals (CI), and a P-value of less than 0.05 was considered to be statistically significant. The pooled results were obtained using the generic inverse of variance method with a random effects model. A total of 21 arms, including 1223 participants (case = 610, and control = 613), reported estradiol concentrations as an outcome measure. The overall results demonstrated that estradiol significantly increased following the administration of DHEA (WMD: 7.02 pg/mL, 95% CI: 5.43, 8.62, P = 0.000). The stratified analyses revealed that the elevation of estradiol concentrations was more pronounced in subjects aged ≥60 years old (WMD: 8.56 pg/mL, 95% CI: 6.97, 10.16, I2 = 94%) and in those receiving DHEA supplements for ≥26 weeks (WMD: 7.30 pg/mL, 95% CI: 6.28, 8.32, I2 = 61%). Moreover, estradiol levels increased significantly with DHEA dosages of 50 mg/day (WMD: 7.75 pg/mL, 95% CI: 9.12, 9.39, I2 = 94%) and when DHEA was prescribed to postmenopausal women (WMD: 7.61 pg/mL, 95% CI: 5.97, 9.24, I2 = 93%). This meta-analysis has provided a comprehensive overview of the effects of DHEA administration on circulating estradiol levels, far beyond the available evidence from different RCTs. Subsequent subgroup analyses revealed that postmenopausal women, females aged 60 years and above, those on DHEA dosages of 50 mg/day and those receiving DHEA for ≥26 weeks registered a more pronounced elevation of the circulating estradiol levels.
Collapse
Affiliation(s)
- Yan Zhu
- Department of Endocrinology, The First Hospital of Nanchang, Nanchang, Jiangxi 330008, China
| | - Lei Qiu
- Department of Oncology, Zhucheng Hospital of Traditional Chinese Medicine, Zhucheng, Shandong 262200, China
| | - Fangfang Jiang
- Department of Endocrinology, The First Hospital of Nanchang, Nanchang, Jiangxi 330008, China
| | - Mihnea-Alexandru Găman
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania; Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Kehinde S Okunade
- Department of Obstetrics & Gynaecology, College of Medicine, University of Lagos/Lagos University Teaching Hospital, PMB 12003, Lagos, Nigeria
| | - Ming Zhang
- Clinical Laboratory, Zhucheng Maternal and Child Health Hospital, Zhucheng, Shandong 262200, China.
| |
Collapse
|
39
|
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
|
40
|
Brunmair J, Bileck A, Stimpfl T, Raible F, Del Favero G, Meier-Menches SM, Gerner C. Metabo-tip: a metabolomics platform for lifestyle monitoring supporting the development of novel strategies in predictive, preventive and personalised medicine. EPMA J 2021; 12:141-153. [PMID: 34188726 PMCID: PMC8192631 DOI: 10.1007/s13167-021-00241-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/14/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND/AIMS Exposure to bioactive compounds from nutrition, pharmaceuticals, environmental contaminants or other lifestyle habits may affect the human organism. To gain insight into the effects of these influences, as well as the fundamental biochemical mechanisms behind them, individual molecular profiling seems to be a promising tool and may support the further development of predictive, preventive and personalised medicine. METHODS We developed an assay, called metabo-tip for the analysis of sweat, collected from fingertips, using mass spectrometry-by far the most comprehensive and sensitive method for such analyses. To evaluate this assay, we exposed volunteers to various xenobiotics using standardised protocols and investigated their metabolic response. RESULTS As early as 15 min after the consumption of a cup of coffee, 50 g of dark chocolate or a serving of citrus fruits, significant changes in the sweat composition of the fingertips were observed, providing relevant information in regard to the ingested substances. This included not only health-promoting bioactive compounds but also potential hazardous substances. Furthermore, the identification of metabolites from orally ingested medications such as metamizole indicated the applicability of this assay to observe specific enzymatic processes in a personalised fashion. Remarkably, we found that the sweat composition fluctuated in a diurnal rhythm, supporting the hypothesis that the composition of sweat can be influenced by endogenous metabolic activities. This was further corroborated by the finding that histamine was significantly increased in the metabo-tip assay in individuals with allergic reactions. CONCLUSION Metabo-tip analysis may have a large number of practical applications due to its analytical power, non-invasive character and the potential of frequent sampling, especially regarding the individualised monitoring of specific lifestyle and influencing factors. The extraordinarily rich individualised metabolomics data provided by metabo-tip offer direct access to individual metabolic activities and will thus support predictive preventive personalised medicine. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13167-021-00241-6.
Collapse
Affiliation(s)
- Julia Brunmair
- Department of Analytical Chemistry, Faculty of Chemistry, University Vienna, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University Vienna, Vienna, Austria
- Joint Metabolome Facility, University and Medical University of Vienna, Vienna, Austria
| | - Thomas Stimpfl
- Joint Metabolome Facility, University and Medical University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Florian Raible
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna, Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Samuel M. Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University Vienna, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University Vienna, Vienna, Austria
- Joint Metabolome Facility, University and Medical University of Vienna, Vienna, Austria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
Rodriguez Paris V, Edwards MC, Aflatounian A, Bertoldo MJ, Ledger WL, Handelsman DJ, Gilchrist RB, Walters KA. Pathogenesis of Reproductive and Metabolic PCOS Traits in a Mouse Model. J Endocr Soc 2021; 5:bvab060. [PMID: 34056500 PMCID: PMC8152184 DOI: 10.1210/jendso/bvab060] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Indexed: 01/02/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common and heterogeneous disorder; however, the etiology and pathogenesis of PCOS are poorly understood and current management is symptom-based. Defining the pathogenesis of PCOS traits is important for developing early PCOS detection markers and new treatment strategies. Hyperandrogenism is a defining characteristic of PCOS, and studies support a role for androgen-driven actions in the development of PCOS. Therefore, we aimed to determine the temporal pattern of development of PCOS features in a well-characterized dihydrotestosterone (DHT)-induced PCOS mouse model after 2, 4, and 8 weeks of DHT exposure. Following 2 weeks of treatment, DHT induced the key PCOS reproductive features of acyclicity, anovulation, and multifollicular ovaries as well as a decrease in large antral follicle health. DHT-treated mice displayed the metabolic PCOS characteristics of increased body weight and exhibited increased visceral adiposity after 8 weeks of DHT treatment. DHT treatment also led to an increase in circulating cholesterol after 2 weeks of exposure and had an overall effect on fasting glucose levels, but not triglycerides, aspartate transaminase (AST) and alanine transaminase (ALT) levels, or hepatic steatosis. These data reveal that in this experimental PCOS mouse model, acyclicity, anovulation, and increased body weight are early features of a developing PCOS phenotype whereas adiposity, impaired glucose tolerance, dyslipidemia, and hepatic steatosis are later developing features of PCOS. These findings provide insights into the likely sequence of PCOS trait development and support the addition of body weight criteria to the early diagnosis of PCOS.
Collapse
Affiliation(s)
- Valentina Rodriguez Paris
- Fertility and Research Centre, School of Women’s & Children’s Health, University of New South Wales Sydney, NSW 2052, Australia
| | - Melissa C Edwards
- Fertility and Research Centre, School of Women’s & Children’s Health, University of New South Wales Sydney, NSW 2052, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales 2139, Australia
| | - Ali Aflatounian
- Fertility and Research Centre, School of Women’s & Children’s Health, University of New South Wales Sydney, NSW 2052, Australia
| | - Michael J Bertoldo
- Fertility and Research Centre, School of Women’s & Children’s Health, University of New South Wales Sydney, NSW 2052, Australia
| | - William L Ledger
- Fertility and Research Centre, School of Women’s & Children’s Health, University of New South Wales Sydney, NSW 2052, Australia
| | - David J Handelsman
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales 2139, Australia
| | - Robert B Gilchrist
- Fertility and Research Centre, School of Women’s & Children’s Health, University of New South Wales Sydney, NSW 2052, Australia
| | - Kirsty A Walters
- Fertility and Research Centre, School of Women’s & Children’s Health, University of New South Wales Sydney, NSW 2052, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales 2139, Australia
| |
Collapse
|
42
|
Pini T, Raubenheimer D, Simpson SJ, Crean AJ. Obesity and Male Reproduction; Placing the Western Diet in Context. Front Endocrinol (Lausanne) 2021; 12:622292. [PMID: 33776921 PMCID: PMC7991841 DOI: 10.3389/fendo.2021.622292] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
There is mounting evidence that obesity has negative repercussions for reproductive physiology in males. Much of this evidence has accumulated from rodent studies employing diets high in fat and sugar ("high fat" or "western" diets). While excessive fats and carbohydrates have long been considered major determinants of diet induced obesity, a growing body of research suggests that the relationships between diet composition and obesity are more complex than originally thought, involving interactions between dietary macronutrients. However, rodent dietary models have yet to evolve to capture this, instead relying heavily on elevated levels of a single macronutrient. While this approach has highlighted important effects of obesity on male reproduction, it does not allow for interpretation of the complex, interacting effects of dietary protein, carbohydrate and fat. Further, the single nutrient approach limits the ability to draw conclusions about which diets best support reproductive function. Nutritional Geometry offers an alternative approach, assessing outcomes of interest over an extended range of dietary macronutrient compositions. This review explores the practical application of Nutritional Geometry to study the effects of dietary macronutrient balance on male reproduction, including experimental considerations specific to studies of diet and reproductive physiology. Finally, this review discusses the promising use of Nutritional Geometry in the development of evidence-based pre-conception nutritional guidance for men.
Collapse
Affiliation(s)
| | | | | | - Angela J. Crean
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
43
|
Gao H, Jiang J, Shi Y, Chen J, Zhao L, Wang C. The LINC00477/miR-128 axis promotes the progression of polycystic ovary syndrome by regulating ovarian granulosa cell proliferation and apoptosis. Reprod Biol Endocrinol 2021; 19:29. [PMID: 33622342 PMCID: PMC7901218 DOI: 10.1186/s12958-021-00718-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/11/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) participate in the pathogenesis of various human diseases. This study aims to investigate the roles of lncRNA LINC00477 in polycystic ovary syndrome (PCOS), especially the impacts of LINC00477 on the proliferation and migration of human granulosa cells and the related mechanisms. METHODS qRT-PCR analysis was performed to examine the expression pattern of LINC00477 in serum samples of PCOS patients as well as PCOS animal models. The effect of LINC00477 on the viability and apoptosis of ovarian granulosa cells was detected by MTT and flow cytometry assays. The correlation between LINC00477 and miR-128 was verified by bioinformatics analysis and dual-luciferase reporter and RNA pull-down assays. Finally, rescue assays were performed to analyze the effects of the LINC00477-miR-128 axis on the biological behaviors of granulosa cells. RESULTS LINC00477 was significantly upregulated in the serum of PCOS patients as well as PCOS mouse models. LINC00477 overexpression inhibited the proliferation and promoted the apoptosis of granulosa cells, whereas knockdown of LINC00477 yielded the opposite effects. Moreover, miR-128 mimics partially abrogated the effect of LINC00477 on granulosa cells. CONCLUSION LINC00477 may function as a ceRNA to inhibit proliferation and apoptosis of granulosa cells by modulating miR-128 expression.
Collapse
Affiliation(s)
- Haijie Gao
- Department of Obstetrics and Gynecology, Shenzhen Hospital, Southern Medical University, No. 1333, Xinhu Road, Shenzhen, 518000, China
- Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Jinna Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Yingying Shi
- Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Jiying Chen
- Department of Obstetrics and Gynecology, Shenzhen Longhua District Central Hospital, Shenzhen, 518000, China
| | - Lijian Zhao
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Chenhong Wang
- Department of Obstetrics and Gynecology, Shenzhen Hospital, Southern Medical University, No. 1333, Xinhu Road, Shenzhen, 518000, China.
| |
Collapse
|
44
|
Affiliation(s)
- Jenny A Visser
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.
| |
Collapse
|