1
|
Lin S, Chen S, Zhang Q. Factors influencing premature ovarian insufficiency: a systematic review and meta-analysis. J OBSTET GYNAECOL 2025; 45:2469331. [PMID: 40013478 DOI: 10.1080/01443615.2025.2469331] [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: 01/08/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Premature ovarian insufficiency (POI) has multiple contributing factors. This study aims to systematically evaluate these factors and their mechanisms, identify high-risk groups, support early intervention, and provide a basis for future research. METHODS Following the PRISMA guidelines, a search was conducted across 11 Chinese and English databases, including PubMed, Embase, and the Cochrane Library, up to January 2024. Observational studies on patients with ovarian insufficiency were included. A meta-analysis using Stata 14 was performed to calculate odds ratios (OR) and 95% confidence intervals (CIs) for factors influencing POI. RESULTS A total of 38 studies with 4,968 cases and 5,158 controls were included. The following factors were identified as risk factors for POI: abnormal menstruation (OR = 2.707, 95% CI: 1.705-4.299), hair dyeing (OR = 4.725, 95% CI: 2.914-7.660), chemical exposure (OR = 3.314, 95% CI: 2.283-4.811), Type A personality (OR = 6.106, 95% CI: 4.696-7.939), survival stress (OR = 3.292, 95% CI: 2.380-4.555), sleep deprivation (OR = 3.340, 95% CI: 2.363-4.721), bad mood (OR = 4.654, 95% CI: 2.783-7.781), smoking history (OR = 2.748, 95% CI: 1.928-3.917), family history of POI (OR = 4.338, 95% CI: 2.422-7.768), history of mumps (OR = 5.802, 95% CI: 3.460-9.730), number of abortions (OR = 2.292, 95% CI: 1.887-2.785), and history of pelvic surgery (OR = 4.836, 95% CI: 3.066-7.628). Physical exercise (OR = 0.270, 95% CI: 0.106-0.692) and vegetable intake (OR = 0.434, 95% CI: 0.337-0.560) were protective factors. Sensitivity analysis and publication bias tests confirmed robustness. CONCLUSIONS POI is related with abnormal menstruation, hair dyeing, chemical exposure, Type A personality, survival stress, sleep deprivation, bad mood, smoking history, family history of POI, history of mumps, number of abortions, and history of pelvic surgery. Physical exercise and vegetable intake may provide protection. These findings inform clinical strategies for early identification and management.
Collapse
Affiliation(s)
- Shuling Lin
- Department of Obstetrics and Gynecology, Puning City Maternal and Child Health Hospital, Guangdong Province, China
| | - Shanyan Chen
- Department of Obstetrics and Gynecology, Puning City Maternal and Child Health Hospital, Guangdong Province, China
| | - Qin Zhang
- Reproductive Medicine Center, Shenzhen Luohu District People's Hospital, Guangdong Province, China
| |
Collapse
|
2
|
Peng Y, Wang Y, Hu J, Wang Z, Liu Y, Ding Z. Trimethylamine N-oxide (TMAO) treatment triggers premature ovarian insufficiency (POI) via the activation of mitochondrial pathway apoptosis in granulosa cells. Free Radic Biol Med 2025; 232:214-230. [PMID: 40054636 DOI: 10.1016/j.freeradbiomed.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Premature ovarian insufficiency (POI) is characterized by follicular development failure or follicular dysplasia, therefore causing the lack of normal ovarian function before 40 years of age. Trimethylamine N-oxide (TMAO) is a metabolite of high choline diet rich in red meat and directly associated with gut microbiota. Correlation of TMAO level with female fertility decline has been shown; however, its mechanism is largely unknown. To unveil the mechanism by which TMAO affects female reproductive function, we established a TMAO-treated mouse model which exhibited the pathological manifestations of POI including increased follicle-stimulating hormone and luteinizing hormone levels, decreased estradiol and anti-Müllerian hormone levels, reduced growing and mature follicles, increased atretic follicles, and decreased fertility. Meanwhile, these mice showed an increased apoptosis ratio and damaged mitochondrial function in granulosa cells, the nursing and supporting cells for oocyte development. Moreover, TMAO treatment significantly elevated oxidative stress and reduced antioxidative capacity in granulosa cells, whereas the antioxidants N-acetylcysteine alleviated such detriment. Mechanism investigation demonstrated that TMAO treatment up-regulated phosphatase and tensin homolog expression levels in granulosa cells, thereby inhibiting the phosphorylation of AKT and subsequently causing high expression of BCL-2-associated X protein, a key molecule in the mitochondria pathway, leading to increased cell apoptosis. Our findings documented the pathological mechanism of TMAO-induced POI, which may provide a potential target for curing POI clinically.
Collapse
Affiliation(s)
- Yuanhong Peng
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingjin Wang
- Department of Obstetrics and Gynecology, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Jiangshan Hu
- Department of Obstetrics and Gynecology, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Zhijie Wang
- Department of Obstetrics and Gynecology, Shanghai Eighth People's Hospital, Shanghai, 200235, China.
| | - Yue Liu
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Zhide Ding
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
3
|
Ammar OF, Liperis G, Sharma K, Ali ZE, Diaz-Gimeno P, Zambelli F, Gould MA, Talaulikar V, Fraire-Zamora JJ. Genomics in diagnosing primary ovarian insufficiency (POI): the door is open, but the path is still to be paved. Hum Reprod 2025:deaf084. [PMID: 40294413 DOI: 10.1093/humrep/deaf084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/08/2025] [Indexed: 04/30/2025] Open
Affiliation(s)
- Omar F Ammar
- Louise, Research and Development Department, Bordeaux, France
- Department of Obstetrics and Gynaecology, College of Medicine, University of Anbar, Ramadi, Iraq
| | - George Liperis
- Westmead Fertility Centre, Institute of Reproductive Medicine, University of Sydney, Westmead, NSW, Australia
- Embryorigin Fertility Centre, Larnaca, Cyprus
| | - Kashish Sharma
- HealthPlus Fertility Center, HealthPlus Network of Specialty Centers, Abu Dhabi, United Arab Emirates
| | - Zoya E Ali
- Research & Development Department, Hertility Health Limited, London, UK
| | - Patricia Diaz-Gimeno
- IVIRMA Global Research Alliance, IVI Foundation-Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | | | - Martine A Gould
- Clinical and Metabolic Genetics Center, Cook Children's Healthcare System, Fort Worth, TX, USA
| | | | | |
Collapse
|
4
|
Hu Z, Gao J, Long P, Quan R, Huang F, Jiang J, Zhang J, Chen J, Xiao H, Huang H. CKAP5 deficiency induces premature ovarian insufficiency. EBioMedicine 2025; 115:105718. [PMID: 40252251 PMCID: PMC12032925 DOI: 10.1016/j.ebiom.2025.105718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is characterized by ovarian dysfunction that develops from diminished ovarian reserve (DOR). The exact aetiology of POI remains poorly understood. This study aims to elucidate the role of CKAP5 in the regulation of ovarian function and fertility. METHODS Bulk RNA sequencing of granulosa cells was conducted in the control group and in the patients with DOR to screen for candidate genes, which were further validated by gene burden analysis in a next-generation sequencing cohort of POI and control individuals. Additionally, ovarian reserve was evaluated in heterozygous Ckap5 knockout mice, alongside the ovarian and oocyte single-cell transcriptome analysis. The regulatory mechanism of CKAP5 was studied through in vivo and in vitro experiments. FINDINGS CKAP5 was identified as a key hub gene associated with ovarian ageing. Heterozygous Ckap5 knockout mice exhibited a POI-like phenotype, characterized by a reduced primordial follicle pool and accelerated follicular atresia. CKAP5 promotes autophagy via ATG7 and simultaneously supports DNA damage repair through the ATM. Finally, a variant in CKAP5 (NM_0001008938.4, c.630 + 7_630 + 11delCAAAA) was identified in patients with POI, resulting in protein truncation and loss of function. INTERPRETATION CKAP5 deficiency induces premature ovarian insufficiency in both humans and mice. FUNDING The National Key R&D Program of China (2017YFC1001100), the National Natural Science Foundation of China (81501248, 81471453 and 81801295), the Health Research Project of Hunan Provincial Health Commission (W20243018), the Science and Technology Innovation Program of Hunan Province (2021RC3031), the National Natural Science Foundation of Hunan Province (2022JJ30066), the Scientific Research Program of Hunan Provincial Health Commission (202205033471 and 21B0058), the Open Research Fund of Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control (HPKL2023013).
Collapse
Affiliation(s)
- Zihao Hu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Reproductive & Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jingping Gao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Panpan Long
- Institute of Reproductive & Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ruping Quan
- Institute of Reproductive & Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Fei Huang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jixuan Jiang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianlin Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hongmei Xiao
- Institute of Reproductive & Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
| | - Hualin Huang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
5
|
Veitia RA, Cowles JD, Caburet S. Reclassifying NOBOX variants in primary ovarian insufficiency cases with a corrected gene model and a novel quantitative framework. Hum Reprod 2025:deaf058. [PMID: 40246288 DOI: 10.1093/humrep/deaf058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 02/19/2025] [Indexed: 04/19/2025] Open
Abstract
STUDY QUESTION How updated expression and genomic data combined with a disease/disorder-specific classification system can be used to correct a gene model for a better evaluation of the pathogenicity of variants found in patients? SUMMARY ANSWER By combining available genomic and transcriptomic data from several species and a quantitative classification framework with primary ovarian insufficiency (POI)-adjusted parameters, we correct the human NOBOX (newborn ovary homeobox) gene model and provide a reclassification of variants previously reported in POI cases. WHAT IS KNOWN ALREADY The NOBOX gene, encoding a gonad-specific transcription factor with a crucial role in early folliculogenesis and considered a major gene involved in POI, is currently described as being expressed as four transcripts, the longest one considered canonical. All the variants identified in POI cases have been evaluated according to this canonical transcript, and the various functional tests have been performed using the corresponding predicted protein. STUDY DESIGN, SIZE, DURATION We refined and corrected the NOBOX gene model using available genomic and RNAseq data in human and 16 other mammalian species. Expression data were selected for tissue specificity, strand specificity, and coverage. The analysis of RNAseq data from different ovarian fetal stages allows for a time-course description of NOBOX isoforms. Literature was scanned to retrieve NOBOX variants reported in POI cases, and NOBOX variants present in ClinVar and GnomAD 4 databases were also retrieved. PARTICIPANTS/MATERIALS, SETTING, METHODS Strand-specific RNAseq data from human fetal ovaries and human adult testes were analysed to infer the correct human NOBOX gene isoforms. The conservation of the gene structure was verified by combining the aligned genomic sequences from 17 mammalian species covering a wide phylogenetic range and the relevant RNAseq data. As changing a gene model implies a reclassification of variants, we set up a quantitative framework with updated variant frequencies from GnomAD4 and POI-adjusted parameters following the American College of Medical Genetics and Genomics/Association for Molecular Pathology (ACMG/AMP) guidelines. Using this framework, we reclassified 44 NOBOX variants reported in POI patients and families, 117 NOBOX variants reported in ClinVar, and 2613 NOBOX variants present in GnomAD4. MAIN RESULTS AND THE ROLE OF CHANCE The corrected NOBOX gene model proposes the invalidation of two transcripts, including the canonical one. The two correct isoforms were present in fetal ovarian samples, and only one was detected in adult testes. Only 14 variants remained as possibly causative for POI. Furthermore, this re-evaluation strongly suggests that NOBOX biallelic variants are the most likely cause of POI. LARGE SCALE DATA Large tables are provided as supplementary data sets on the Zenodo repository. LIMITATIONS, REASONS FOR CAUTION The proposed gene model is robust but relies on available transcriptomic data covering a range of time points and tissues. Our scoring system was manually adjusted and other laboratories can implement it with different parameters. WIDER IMPLICATIONS OF THE FINDINGS For the NOBOX variants that cannot be considered pathogenic or causative anymore, the genome/exome sequencing data of the corresponding patients should be reanalysed. Furthermore, the functional studies performed using the obsolete coding sequence should be reconsidered. The corrected gene model should be taken into account when evaluating novel NOBOX variants identified in POI patients. Our results highlight the importance of the careful assessment of the most updated expression data for validating a gene model, enabling a correct evaluation of the pathogenicity of variants found in patients. The proposed quantitative framework developed here can be used for the classification of variants in other genes underlying POI. Furthermore, the global approach based on quantitatively adjusting the ACMG/AMP guidelines could be extended to other inherited pathologies. STUDY FUNDING/COMPETING INTEREST(S) This project was not funded. All the authors have no conflict of interest to disclose.
Collapse
Affiliation(s)
- Reiner A Veitia
- Department of Life Sciences, Université Paris Cité, CNRS, Institut Jacques Monod, CNRS UMR7592, Paris, France
- Department of Life Sciences, Université Paris Saclay, Gif-sur-Yvette, France
- Institut de Biologie François Jacob, CEA, Fontenay aux Roses, France
| | - Jamie D Cowles
- Department of Life Sciences, Université Paris Cité, CNRS, Institut Jacques Monod, CNRS UMR7592, Paris, France
| | - Sandrine Caburet
- Department of Life Sciences, Université Paris Cité, CNRS, Institut Jacques Monod, CNRS UMR7592, Paris, France
| |
Collapse
|
6
|
Hassan RM, Elzayat EM, Eid JI, Abdelgayed SS, Hosney M. Protective effects of Moringa oleifera leaf extract against cyclophosphamide-induced ovarian dysfunction and follicular loss in rats. Tissue Cell 2025; 95:102916. [PMID: 40233669 DOI: 10.1016/j.tice.2025.102916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
The current study aims to determine whether Moringa oleifera (M. oleifera) leaf extract can reverse cyclophosphamide (CP)-induced ovarian dysfunction and follicle loss in rats, potentially through antioxidant or anti-inflammatory pathways. Female rats were divided into four experimental groups: (1) negative control (administrated distilled water), (2) premature ovarian failure (POF) model group (induced by a single intraperitoneal dose of CP), (3) M. Oleifera extract alone, and (4) M. oleifera + CP. CP induced multiple effects on the ovaries, including hormonal imbalances (increased FSH and decreased E2 levels), oxidative stress (elevated serum MDA and NO levels), altered gene expression (upregulated TNF-α and downregulated TGF-β), and histological changes (follicular atresia and stromal hyperplasia). Pretreatment with M. oleifera successfully mitigated CP-induced oxidative and inflammatory changes, as well as ovarian tissue damage, but failed to reverse serum hormonal imbalances. These findings demonstrate the protective potential of M. oleifera leaf extract against CP-induced ovarian toxicity, likely mediated by the synergistic antioxidant, anti-inflammatory, and organ-protective properties of its bioactive components.
Collapse
Affiliation(s)
- Rehab Mohamed Hassan
- Department of Biotechnology, Faculty of Biotechnology, MUST University, Giza, Egypt
| | - Emad M Elzayat
- Department of Biotechnology, Faculty of Science, Cairo University, Giza, Egypt
| | - Jehane I Eid
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt.
| | - Sherein S Abdelgayed
- Pathobiology Department, College of Veterinary Medicine, Tuskegee University, AL36088, USA; Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Mohamed Hosney
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
7
|
Liang Y, Ou J, Fu J, Wang Y, Li Y, Li J, Yi Y. Smoking, Genetic Susceptibility and Early Menopause: Unveiling Biological Mechanisms and Potential Therapy Targets. BJOG 2025; 132:625-637. [PMID: 39727065 DOI: 10.1111/1471-0528.18052] [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: 09/25/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE To explore the association between smoking, genetic susceptibility and early menopause (EM) and clarify the potential mechanisms underlying this relationship. DESIGN An observational and Transcriptome-wide association analysis (TWAS) study. SETTING UK Biobank and public summary statistics. POPULATION 139 869 women with full baseline and menopause data, and no gynaecological surgery history. METHODS Adjusted modified Poisson regression models were developed to determine the smoking and genetic risk effects on EM. TWAS was used to identify gene expression between smoking and EM, with Mendelian randomisation (MR) to infer causality. Enrichment analysis explored regulatory networks of transcription factors, microRNAs and potential therapeutic targets. Small molecule drugs were predicted using drug-gene interaction analysis. MAIN OUTCOME MEASURES EM prevalence and common gene expression patterns. RESULTS Women with over 30 pack-years of smoking had about 1.5 times higher EM risk, with RRs of 1.39 (95%CI, 1.23-1.56), 1.45 (1.33-1.59) and 1.45 (1.36-1.55) in the low, intermediate and high genetic risk groups. TWAS identified hub genes such as IMMP2L, BMPR2 and HMGN1. MR confirmed daily cigarette consumption as a causal factor in early menopause. Several potential therapeutic targets (e.g., SP600125, INCB18424 and ruxolitinib) were identified. CONCLUSIONS Smoking reduction significantly lowered the risk of EM. Hub genes and therapeutic targets identified provided new avenues for mitigating harmful effects of smoking.
Collapse
Affiliation(s)
- Yuhang Liang
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
- Bioinformatics Center, Furong Laboratory, National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Ou
- Department of Gyneacology, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Fu
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha, China
| | - Yijing Wang
- Bioinformatics Center, Furong Laboratory, National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yanping Li
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Women's Reproductive Health in Hunan Province, Changsha, China
| | - Jinchen Li
- Bioinformatics Center, Furong Laboratory, National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Yi
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
- Bioinformatics Center, Furong Laboratory, National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
8
|
Ding Z, Shao G, Li M. Targeting autophagy in premature ovarian failure: Therapeutic strategies from molecular pathways to clinical applications. Life Sci 2025; 366-367:123473. [PMID: 39971127 DOI: 10.1016/j.lfs.2025.123473] [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: 09/11/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Premature ovarian failure (POF) is a condition where the ovaries lose their function before the age of 40, leading to significant impacts on reproductive health and overall well-being. Current treatment options are limited and often ineffective at restoring ovarian function. This review explores the role of autophagy- a cellular process that helps maintain homeostasis by recycling damaged components-in the development and potential treatment of POF. Autophagy is crucial for the survival of follicle cells and can be disrupted by various stressors associated with POF, such as oxidative damage and mitochondrial dysfunction. We review several key molecular pathways involved in autophagy, including the PI3K/AKT/mTOR, PINK1-Parkin, JAK2/STAT3, MAPK and AMPK/FOXO3a pathways, which have been implicated in POF. Each pathway offers unique insights into how autophagy can be modulated to counteract POF-related damage. Additionally, we discuss emerging therapeutic strategies that target these pathways, including chemical compounds, peptides, hormones, RNA therapy, extracellular vesicles and traditional Chinese medicine. These approaches aim to restore autophagic balance, promote follicle survival and improve ovarian function. By targeting autophagy, new treatments may offer hope for better management and potential reversal of POF, thus improving the quality of life for affected individuals.
Collapse
Affiliation(s)
- Ziwen Ding
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Genbao Shao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Mingyang Li
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
9
|
He X, Chang X, Zhuang S, Liu J, Wang Y, Qin Y, Guo T. Genome-wide DNA methylation profile and predictive biomarkers in premature ovarian insufficiency. J Genet Genomics 2025; 52:596-599. [PMID: 39426591 DOI: 10.1016/j.jgg.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Affiliation(s)
- Xinmiao He
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Xinyue Chang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Shuning Zhuang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Jianing Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Yuteng Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Yingying Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Ting Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
10
|
Stokes G, Herath M, Samad N, Trinh A, Milat F. 'Bone Health-Across a Woman's Lifespan'. Clin Endocrinol (Oxf) 2025; 102:389-402. [PMID: 39871618 PMCID: PMC11874200 DOI: 10.1111/cen.15203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/17/2024] [Accepted: 01/09/2025] [Indexed: 01/29/2025]
Abstract
Despite a high burden of osteoporosis and minimal trauma fractures worldwide, there is still a treatment gap in timely diagnosis and optimal treatment. There is also a lack of international consensus and guidelines on the management of bone fragility in premenopausal women. This review article provides an overview of the current understanding of factors impacting women's bone health across the adult lifespan, as well as dilemmas in the diagnosis, assessment and management of osteoporosis in premenopausal and postmenopausal women, premature ovarian insufficiency and bone health following breast cancer.
Collapse
Affiliation(s)
- Gabrielle Stokes
- Centre for Endocrinology & Metabolism, Hudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of MedicineSchool of Clinical SciencesMonash UniversityClaytonVictoriaAustralia
- Department of EndocrinologyMonash HealthClaytonVictoriaAustralia
| | - Madhuni Herath
- Centre for Endocrinology & Metabolism, Hudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of MedicineSchool of Clinical SciencesMonash UniversityClaytonVictoriaAustralia
- Department of EndocrinologyMonash HealthClaytonVictoriaAustralia
| | - Navira Samad
- Centre for Endocrinology & Metabolism, Hudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of MedicineSchool of Clinical SciencesMonash UniversityClaytonVictoriaAustralia
- Department of EndocrinologyMonash HealthClaytonVictoriaAustralia
| | - Anne Trinh
- Centre for Endocrinology & Metabolism, Hudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of MedicineSchool of Clinical SciencesMonash UniversityClaytonVictoriaAustralia
- Department of EndocrinologyMonash HealthClaytonVictoriaAustralia
| | - Frances Milat
- Centre for Endocrinology & Metabolism, Hudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of MedicineSchool of Clinical SciencesMonash UniversityClaytonVictoriaAustralia
- Department of EndocrinologyMonash HealthClaytonVictoriaAustralia
| |
Collapse
|
11
|
Sugianto SRS, Webber L, Safdar Husain F, Viardot-Foucault V, Nadarajah S, Lim JY, Tan ES, Yong TT, Puvanendran R. Premature ovarian insufficiency: When ovaries retire early. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2025; 54:178-191. [PMID: 40178424 DOI: 10.47102/annals-acadmedsg.2024227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Introduction Premature ovarian insufficiency (POI) refers to loss of ovarian activity before the age 40 years. POI has significant detrimental effects on health (infertility, cardiovascular diseases, type 2 diabetes, reduced bone density, dementia), well-being and longevity. This summary is a practical toolkit for health-care professionals (HCPs) looking after women with POI. Method A workgroup comprising specialists in gynaecology, reproductive medicine, endocrinology, genetics and family medicine reviewed relevant guidelines and literature on POI to establish recom-mendations for the diagnosis and management of POI in Singapore. Results A summary to assist HCPs manage POI was produced, outlining: (1) the aetiology and conse-quences of POI; (2) making the diagnosis; (3) hormone therapy (HT) prescribing options including for those with additional medical conditions; (4) counselling women with POI about HT; and (5) long-term management of POI. Conclusion Timely diagnosis and management of POI is vital to prevent long-term adverse consequences, except infertility. HT is the mainstay of treatment and there are no alternatives as effective. Contraindications are very few; estrogen-sensitive cancer is the main contraindication, and caution in prescribing may be needed with established coexisting cardiovascular disease. Estrogen dosage is higher than when treating normal menopause, and as a result, the patient might require more progestogen for endometrial protection. Minimising cardiovascular risk factors by following a healthy lifestyle is important. POI is a significant public health issue and it is imperative that women have affordable access to appropriate HT. Large-scale research on POI in Asian women is needed.
Collapse
Affiliation(s)
| | - Lisa Webber
- Department of Obstetrics and Gynaecology, Singapore General Hospital, Singapore
| | - Farah Safdar Husain
- Department of Family Medicine, KK Women's and Children's Hospital, Singapore
| | | | - Sadhana Nadarajah
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore
| | - Jiin Ying Lim
- Genetics Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore
| | - Ee Shien Tan
- Genetics Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore
| | - Tze Tein Yong
- Department of Obstetrics and Gynaecology, Singapore General Hospital, Singapore
| | | |
Collapse
|
12
|
Ramezani Tehrani F, Mousavi M, Saei Ghare Naz M, Noroozzadeh M, Azizi F, Farahmand M. Endogenous Estrogen Exposure and Hypertension Risk; A Population-based Cohort Study With About 2 Decades of Follow-up. J Clin Endocrinol Metab 2025; 110:e1125-e1133. [PMID: 38723162 DOI: 10.1210/clinem/dgae316] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Indexed: 03/19/2025]
Abstract
CONTENT The impact of endogenous estrogen exposure (EEE) on hypertension (HTN) incidence has not been investigated yet. OBJECTIVE This study aimed to evaluate HTN incidence in women with different endogenous estrogen durations. METHODS Information was gathered from the Tehran Lipid and Glucose Study to conduct current research. At the initiation of the study, 4463 postmenarche normotensive women, including 3599 premenopausal and 864 menopausal women, were included. EEE was calculated for each woman, and they were followed up for the HTN event. According to the EEE, the hazard ratios and 95% confidence intervals (CI) for the HTN event were presented using Cox proportional hazards regression models (unadjusted and adjusted). RESULTS The median (interquartile range) of follow-up (between menarche and the date of HTN incidence or last follow-up) was 33.2 (25.1, 42.3) years. The event of menopause occurred in 31.8% of participants. The unadjusted model's findings illustrated that the EEE z-score was inversely associated with HTN incidence in postmenarcheal women [unadjusted hazard ratio (HR) .47, 95% CI .44-.50], meaning that the risk of HTN decreased by 53% for every 1-SD rise in the EEE z-score. After adjusting for potential confounders, the results showed no statistically significant changes (adjusted HR .46, 95% CI .43-.49). In participants with prehypertension at baseline, the hazard of HTN decreased by 56% per 1-SD rise in the EEE z-score. CONCLUSION This longitudinal study demonstrated the protective effect of a longer EEE duration on HTN risk, even among those with prehypertension status.
Collapse
Affiliation(s)
- Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran
| | - Maryam Mousavi
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran
| | - Marzieh Saei Ghare Naz
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran
| | - Mahsa Noroozzadeh
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran
| | - Maryam Farahmand
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran
| |
Collapse
|
13
|
Ma W, Ye S, Tian L, Liu M, Wang R, Yang X, Wang M, Fu F, Ren W, Dang L, Wang T, Wang W, Wang S, Sun Y, Li Y. Association of LHCGR rs2293275 genotype with ovarian aging in Chinese women: a multicenter population-based study. Reprod Biol Endocrinol 2025; 23:41. [PMID: 40089715 PMCID: PMC11909885 DOI: 10.1186/s12958-025-01375-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/02/2025] [Indexed: 03/17/2025] Open
Abstract
OBJECTIVE To evaluate the association between the LHCGR rs2293275 (N312S) genotype and ovarian aging phenotypes in Han Chinese women, focusing on diminished ovarian reserve (DOR) and primary ovarian insufficiency (POI). STUDY DESIGN This multicenter population-based study included 1,240 women aged 18-40 years diagnosed with DOR (n = 711) or POI (n = 529), alongside 72,846 ethnically and regionally matched controls from the Han Chinese Genomes Database (PGG.Han). Genotyping of rs2293275 was performed, and clinical data (menstrual history, hormonal profiles, maternal menopause age, and ART outcomes) were analyzed. MAIN RESULTS The AA genotype frequency in the ovarian aging cohort (1.85%) was significantly higher than in the general Han population (0.62%, OR 3.04, 95% CI 1.99-4.64, p < 0.001). AA carriers exhibited earlier POI diagnosis (25.5 ± 6.4 vs. 32.0 ± 5.1 years in GG carriers, p < 0.001) and maternal menopause (41.6 ± 3.3 vs. 47.8 ± 4.1 years, p < 0.001). In controlled ovarian stimulation cycles, AA carriers demonstrated reduced ovarian sensitivity (OSI: 3.59 vs. 1.21 in GG, p = 0.019) despite comparable gonadotropin doses. CONCLUSIONS The LHCGR rs2293275 AA genotype is strongly associated with accelerated ovarian aging in Han Chinese women, highlighting its potential as a biomarker for early identification of high-risk individuals. While these findings underscore genetic contributions to ovarian dysfunction, further mechanistic studies are needed to establish causality and optimize clinical translation. TRIAL REGISTRATION NUMBER ClinicalTrials.gov NCT05665010, registered on 2022-11-30.
Collapse
Affiliation(s)
- Wenqing Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuangmei Ye
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lifeng Tian
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Women's Reproductive Health, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, 330000, China
| | - Min Liu
- Changsha Maternal and Child Health Hospital, Changsha, 410000, China
| | - Rui Wang
- Henan Maternal and Child Health Hospital, Zhengzhou, 450000, China
| | - Xuezhou Yang
- Department of Reproductive Center, Affiliated Hospital of Hubei, Xiangyang Central Hospital, University of Arts and Science, Xiangyang, 441000, China
| | - Man Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fangfang Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wu Ren
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lei Dang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tian Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenwen Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Sun
- Fujian Maternal and Child Health Hospital, Fuzhou, 350001, China.
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
14
|
Zhang Z, Liu P, Xue X, Zhang Z, Wang L, Jiang Y, Zhang C, Zhou H, Lv S, Shen W, Yang S, Wang F. The role of platelet-rich plasma in biomedicine: A comprehensive overview. iScience 2025; 28:111705. [PMID: 39898035 PMCID: PMC11787504 DOI: 10.1016/j.isci.2024.111705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025] Open
Abstract
Biomedicine has seen significant advancements in the 21st century, with platelet-rich plasma (PRP) playing a crucial role in clinical practice. This blood derivative, enriched with platelet components, has shown great potential for promoting tissue repair and regeneration. Its wide range of applications and the presence of anti-inflammatory and growth-promoting factors make it a valuable tool in the field of biomedicine. The exploration of PRP in clinical settings has been gaining momentum. Despite its cost-effectiveness, safety, and therapeutic efficacy, the widespread clinical adoption of PRP has been hindered by the absence of consistent preparation standards and standardized treatment protocols. This article provides a comprehensive analysis of the clinical uses, physiological roles, molecular mechanisms, and preparation techniques of PRP in biomedicine. The aim is to offer a thorough understanding of the potential applications and benefits of PRP in medical practice.
Collapse
Affiliation(s)
- Zhixin Zhang
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
- Graduate School of Medicine, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
| | - Peng Liu
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
- Graduate School of Medicine, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
| | - Xinmiao Xue
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
- Graduate School of Medicine, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
| | - Zhiyu Zhang
- School of Physics and Optoelectronic Engineering Xidian University, Xi’an 710071, China
| | - Li Wang
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
- Graduate School of Medicine, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
| | - Yvke Jiang
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
- Graduate School of Medicine, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
| | - Chi Zhang
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
- Graduate School of Medicine, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
| | - Hanwen Zhou
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
| | - Shuhan Lv
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
| | - Weidong Shen
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
- Graduate School of Medicine, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
| | - Shiming Yang
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
- Graduate School of Medicine, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
| | - Fangyuan Wang
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Chinese PLA Medical School, Beijing 100853, China
- State Key Laboratory of Hearing and Balance Science, Beijing 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing 100853, China
| |
Collapse
|
15
|
Crankshaw TL, Manzini-Matebula N. The continuum of menstrual health through the life course. Lancet Glob Health 2025; 13:e183-e184. [PMID: 39890216 DOI: 10.1016/s2214-109x(24)00519-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 02/03/2025]
Affiliation(s)
- Tamaryn L Crankshaw
- Health Economics and HIV and AIDS Research Division, University of KwaZulu-Natal, Durban, South Africa.
| | | |
Collapse
|
16
|
Panay N, Anderson RA, Bennie A, Cedars M, Davies M, Ee C, Gravholt CH, Kalantaridou S, Kallen A, Kim KQ, Misrahi M, Mousa A, Nappi RE, Rocca WA, Ruan X, Teede H, Vermeulen N, Vogt E, Vincent AJ. Evidence-based guideline: Premature Ovarian Insufficiency. Fertil Steril 2025; 123:221-236. [PMID: 39652037 DOI: 10.1016/j.fertnstert.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 11/06/2024] [Indexed: 02/04/2025]
Abstract
STUDY QUESTION How should premature/primary ovarian insufficiency (POI) be diagnosed and managed, based on the best available evidence from published literature? SUMMARY ANSWER The current guideline provides 145 recommendations on symptoms, diagnosis, causation, sequelae and treatment of POI. WHAT IS KNOWN ALREADY Premature ovarian insufficiency (POI) presents a significant challenge to women's health, with far-reaching implications, both physically and emotionally. The potential implications include adverse effects on quality of life; fertility; and bone, cardiovascular and cognitive health. Although hormone therapy (HT) can mitigate some of these effects, many questions still remain regarding the optimal management of POI. STUDY DESIGN, SIZE, DURATION The guideline was developed according to the structured methodology for development of ESHRE guidelines. Key questions were determined by a group of experts and informed by a scoping survey of women and health care professionals. Literature searches and assessment were then performed. Papers published up to January 30th, 2024, and written in English were included in the guideline. An integrity review was conducted for the randomised controlled trials (RCTs) on POI included in the guideline. PARTICIPANTS/MATERIALS, SETTING, METHODS Based on the collected evidence, recommendations were formulated and discussed within the guideline development group until consensus was reached. Women with lived experience of POI informed the recommendations in general, and particularly on those on provision of care. A stakeholder review was organised after finalisation of the draft. The final version was approved by the guideline development group and the ESHRE Executive Committee. MAIN RESULTS AND THE ROLE OF CHANCE New data indicate a higher prevalence of POI, 3.5%, than was previously thought. This guideline aims to help health care professionals to apply best practice care for women with POI. The recent update of the POI guideline covers 40 clinical questions on diagnosis of the condition, the different sequelae, including bone, cardiovascular, neurological and sexual function, fertility and general well-being, and treatment options, including hormone therapy. The list of clinical questions was expanded from the previous iteration of the guideline (2015) based on the scoping survey and appreciation of emerging knowledge of POI. Questions were added on the role of anti-Müllerian hormone (AMH) in the diagnosis of POI, fertility preservation, muscle health, and specific considerations for HT in iatrogenic POI. Additionally, the topic on complementary treatments was extended with specific focus on non-hormonal treatments and lifestyle management options. Significant changes from the previous 2015 guideline include the recommendations that only one elevated FSH >25 IU is required for diagnosis of POI and guidance that AMH testing, repeat FSH measurement and/or AMH may be required where there is diagnostic uncertainty. Recommendations were also updated regarding genetic testing, estrogen doses and regimens, use of the combined oral contraceptive and testosterone therapy. Women with lived experience of POI informed the recommendations on provision of care. LIMITATIONS, REASONS FOR CAUTION The guideline describes different management options, but it must be acknowledged that for most of these options, supporting evidence is limited for POI. WIDER IMPLICATIONS OF THE FINDINGS The guideline provides health care professionals with clear advice on best practice in POI care, based on the best evidence currently available. In addition, a list of research recommendations is provided to guide further studies in POI. STUDY FUNDING/COMPETING INTEREST(S) The guideline was developed and funded by ESHRE, American Society for Reproductive Medicine (ASRM), Centre for Research Excellence in Women's Health in Repoduction Life (CRE-WHiRL) and International Menopause Society (IMS), covering expenses associated with the guideline meetings, literature searches and dissemination of the guideline. The guideline group members did not receive payments. N.P. declared grants from Bayer Pharma (research and consultancy), and NIHR - research POISE; consulting fees from Abbott, Astellas, Bayer, Besins, Lawley, Mithra, Theramex, Viatris; honoraria from Astellas, Bayer, Besins, Gedeon Richter, Theramex, Viatris; support for attending meetings and/or travel from Astellas, Bayer, Theramex, Viatris; President, International Menopause Society, Medical Advisory Committee member, British Menopause Society, Patron Daisy Network. A.J.V. declared grants from Amgen Australia, Australian NHMRC, and Australian MRFF; consulting fees from IQ Fertility; honoraria from the Australasian Menopause Society; participation on a Data Safety Monitoring Board or Advisory Board of Astellas; Board Member of the International Menopause Society (2020 to current) and Past president of the Australasian Menopause Society (2017-2019); R.A.A. declared grants from Roche (Research support, to institution), and participation on a Data Safety Monitoring Board of Bayer. M.C. declared grants from NHI; payments or honoraria from Up-to-Date (as editor/reviewer); Board Member of American Society of Reproductive Medicine, and of American Gynecological and Obstetrical Society. M.D. declared (NIHR - HTA Reference Number: NIHR133461; NIHR - HTA Reference Number: NIHR128757; Action Medical Research and Borne: GN2818); consulting fees from a small personal medical practice, support for attending meetings and/or travel from ESHRE, Bayer and UCLH special Trustees; Participation on the Advisory Board from the British Menopause Society, UKSTORE project, the Progress Educational Trust, and the Turner Syndrome Support Society UK; Leadership or fiduciary roles in the British Fertility Society (Trustee), Elizabeth Garrett Anderson Hospital Charity (chair of Trustees), and the Essex Wynter charitable trust (Trustee). C.E. declared being Chair of a SIG from the Royal Australian College of General Practitioners Integrative Medicine Specific Interest Group and Program Lead for Next Practice Western Sydney Integrative Health. C.H.G. declared grants from Novo Nordisk Foundation (Nos. NNF15OC0016474 and NNF20OC0060610), sygesikringen danmark (No 2022-0189), and the Independent Research Fund Denmark (Nos. 0134-00406 and 0134-00130B); consulting fees from Novo Nordisk, Merck, and Astra Zeneca. S.K. declared grants from Roche diagnostics. A.K. declared grants from NIH R01 5R01HD101475; consulting fees as Medical Reviewer for Flo and for Healthline; honoraria as Medical Consultant for Summus; support for attending meetings from the Reproductive Scientist Development Program; Society for Reproductive Investigation Council Member and Society for Assisted Reproduction Registry / Validation Chair; R.N. declared consulting fees from Astellas, Bayer Pharma, Besins Healthcare, Fidia, Theramex; honoraria from Abbott, Astellas, Exeltis, Fidia, Gedeon Richter, Merck & Co, Novo Nordisk, Shionogi Limited, Theramex, Viatris; payment for expert testimony from Vichy Laboratories; Participation in Data Safety Monitoring Board of Advisory board from Astellas and Bayer Healthcare; President elect of the International Menopause Society (IMS). H.T. declared a grant from NHMRC Centre for Research Excellence for women's health in reproductive life. A.B. declared being chair of the Daisy Network Charity. The other authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- Nick Panay
- Queen Charlotte's and Chelsea Hospital, Imperial College London, UK.
| | - Richard A Anderson
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, UK
| | | | - Marcelle Cedars
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, School of Medicine, San Francisco, CA, USA
| | - Melanie Davies
- Reproductive Medicine Unit, University College London Hospital, London, UK
| | - Carolyn Ee
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Claus H Gravholt
- Department of Endocrinology, Aarhus University Hospital, Aarhus, Denmark
| | - Sophia Kalantaridou
- Third Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Athens, Greece
| | - Amanda Kallen
- Division of Reproductive Endocrinology and Infertility, University of Vermont Larner College of Medicine, Burlington, VT, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Kimberly Q Kim
- RESOLVE: The National Infertility Association, McLean, VA, USA
| | - Micheline Misrahi
- Université Paris Saclay, Faculté de Médecine, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Aya Mousa
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Clayton, Australia
| | - Rossella E Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS San Matteo Foundation, Pavia, Italy
| | - Walter A Rocca
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Beijing, China
| | - Helena Teede
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Clayton, Australia
| | | | - Elinor Vogt
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Amanda J Vincent
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Clayton, Australia
| |
Collapse
|
17
|
Kong D, Nie Y, He H, Guo H. Investigating the role and mechanisms of bisphenol compounds in premature ovarian insufficiency using computational biology and bioinformatics. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117789. [PMID: 39874714 DOI: 10.1016/j.ecoenv.2025.117789] [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: 09/28/2024] [Revised: 01/08/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025]
Abstract
Premature Ovarian Insufficiency refers to the premature decline in ovarian function before the age of 40, resulting in menstrual irregularities or complete cessation of menstruation, and affecting fertility. Widely used bisphenol compounds may have potential health effects, including premature ovarian insufficiency (POI). This study employs computational biology and bioinformatics to investigate the effects of bisphenols (BPs) on POI. Using bioinformatics tools, we identified potential target genes related to both bisphenols and POI, and conducted functional enrichment analysis. Further, we calculated differentially expressed genes for POI, extracted core networks, and explored immune function and screened core genes. Molecular docking and molecular dynamics simulations were used to explore the stable binding between bisphenols and core POI genes. Our results constructed a protein network of 56 potential target genes and extracted core subnetworks. Functional enrichment analysis indicated that estrogen metabolism, estrogen receptor pathways, steroid hormone metabolism, and carbohydrate and lipid metabolism may be involved in the process of BPs-induced POI. The differentially expressed genes obtained through further screening, CYP1A1 and CYP19A1, were subjected to molecular docking and dynamics simulations to reveal the mechanism by which bisphenols participate in estrogen metabolism through their stable binding. Our findings underscore the role of bisphenols in inducing POI and the potential mechanisms involved, providing new directions for further epidemiological and molecular biological research into this regulatory process.
Collapse
Affiliation(s)
- Deshui Kong
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
| | - Yufei Nie
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
| | - Haojie He
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
| | - Hongyan Guo
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
| |
Collapse
|
18
|
Zou Y, Li Z, Lin Y, Zheng Y, Liu Z, Li Y, Huang L, Chen Z, Zhu L. Shanyao regulates the PI3K/AKT/P21 pathway to promote oogonial stem cell proliferation and stemness restoration to alleviate premature ovarian insufficiency. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119168. [PMID: 39615771 DOI: 10.1016/j.jep.2024.119168] [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: 10/12/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shanyao (SY, yam, Rhizoma Dioscoreae, the dried rhizome of Dioscorea oppositifolia L.) was recorded in the Chinese pharmacopoeia and was often used in the treatment of premature ovarian insufficiency (POI). AIM OF STUDY To evaluate the efficacy of shanyao in cyclophosphamide (CTX)-induced POI and explore its potential mechanism of action. MATERIAL AND METHODS We employed network pharmacology, Liquid Chromatograph Mass Spectrometer (LC-MS), and molecular docking methods to identify active compounds and core targets, and predict the mechanism of shanyao for treating POI. The mechanism was subsequently validated through a series of experiments. Female Sprague-Dawley (SD) rats were randomly divided into five groups: control (CON), model, estradiol valerate (EV), low-dose shanyao, and high-dose shanyao. An experimental rat model of POI was established using cyclophosphamide and treated with either shanyao or EV for a duration of two months. We assessed the efficacy of shanyao in vivo through methods such as weighing, Enzyme-linked Immunosorbent Assay (ELISA), and Hematoxylin and Eosin (H&E) staining. Oogonial stem cells (OSCs) were isolated, after modeling, treated them with a serum containing either shanyao or EV. Using methods such as CCK8 assay, immunofluorescence staining, flow cytometry (FCM) analysis, and Western blot analysis to verify the mechanism of shanyao in treating POI. RESULTS In this study, we found that after treatment with shanyao, the general condition of POI rats was improved, body weight and the ratio of ovarian weight to body weight were increased, FSH, E2 and AMH levels were improved, primary follicles and preantral follicles were significantly increased, atretic follicles were decreased. However, the number of antral follicles and fresh corpus luteum was no statistical difference. We identified 10 active compounds of shanyao that act on 220 target genes, 176 of which are associated with POI. Denudatin B and Kadsurenone were finally identified as core components. Through topological analysis, 18 key targets were selected, and ultimately PI3K, CCND1, and CDK4 were identified as core targets. Molecular docking results showed that core components had good binding energy with core targets. The results of GO and KEGG enrichment analysis mainly focus on cell cycle regulation and PI3K/AKT signaling pathway. A series of molecular biology experiments confirmed that after shanyao treatment, the phosphorylation level of PI3K and AKT in POI rats were increased, P21 was inhibited, PI3K/AKT/P21 signaling pathway was activated, and the expression levels of CCND1 and CDK4 were increased. At the same time, the expression of Oct4, fragilis and Mvh of ovarian stem cells was up-regulated. CONCLUSION The active compounds of shanyao can regulate the PI3K/AKT/P21 signaling pathway, promote the proliferation of oogonial stem cells, stemness restoration, and delay ovarian aging. This study provides valuable insights into shanyao treatment for POI.
Collapse
Affiliation(s)
- Yuxin Zou
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Zuang Li
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yuewei Lin
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yunling Zheng
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Ziyan Liu
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yucheng Li
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Liuqian Huang
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Zhuoting Chen
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Ling Zhu
- Department of Gynecology, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
19
|
Kline BL, Siddall NA, Wijaya F, Stuart CJ, Orlando L, Bakhshalizadeh S, Afkhami F, Bell KM, Jaillard S, Robevska G, van den Bergen JA, Shahbazi S, van Hoof A, Ayers KL, Hime GR, Sinclair AH, Tucker EJ. Functional characterization of human recessive DIS3 variants in premature ovarian insufficiency†. Biol Reprod 2025; 112:102-118. [PMID: 39400047 PMCID: PMC11736438 DOI: 10.1093/biolre/ioae148] [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: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/16/2024] [Indexed: 10/15/2024] Open
Abstract
Premature ovarian insufficiency (POI) is characterized by the loss or complete absence of ovarian activity in women under the age of 40. Clinical presentation of POI varies with phenotypic severity ranging from premature loss of menses to complete gonadal dysgenesis. POI is genetically heterogeneous with >100 causative gene variants identified thus far. The etiology of POI varies from syndromic, idiopathic, monogenic to autoimmune causes the condition. Genetic diagnoses are beneficial to those impacted by POI as it allows for improved clinical management and fertility preservation. Identifying novel variants in candidate POI genes, however, is insufficient to make clinical diagnoses. The impact of missense variants can be predicted using bioinformatic algorithms but computational approaches have limitations and can generate false positive and false negative predictions. Functional characterization of missense variants, is therefore imperative, particularly for genes lacking a well-established genotype:phenotype correlation. Here we used whole-exome sequencing (WES) to identify the first case of a homozygous missense variant in DIS3 (c.2320C > T; p.His774Tyr) a critical component of the RNA exosome in a POI patient. This adds to the previously described compound heterozygous patient. We perform the first functional characterization of a human POI-associated DIS3 variant. A slight defect in mitotic growth was caused by the variant in a Saccharomyces cerevisiae model. Transgenic rescue of Dis3 knockdown in Drosophila melanogaster with human DIS3 carrying the patient variant led to aberrant ovarian development and egg chamber degeneration. This supports a potential deleterious impact of the human c.2320C > T; p.His774Tyr variant.
Collapse
Affiliation(s)
- Brianna L Kline
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Grattan Street, Parkville, VIC 3010, Melbourne, Australia
| | - Nicole A Siddall
- Department of Anatomy and Physiology, The University of Melbourne, Grattan Street, Parkville, VIC 3010, Melbourne, Australia
| | - Fernando Wijaya
- Department of Anatomy and Physiology, The University of Melbourne, Grattan Street, Parkville, VIC 3010, Melbourne, Australia
| | - Catherine J Stuart
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center at Houston, 7000 Fannin, Suite 1706, Houston, TX 77030, USA
| | - Luisa Orlando
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center at Houston, 7000 Fannin, Suite 1706, Houston, TX 77030, USA
| | - Shabnam Bakhshalizadeh
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Grattan Street, Parkville, VIC 3010, Melbourne, Australia
| | - Fateme Afkhami
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran Province, Tehran, Jalal Al Ahmad St, P9CJ+HC9, Iran
| | - Katrina M Bell
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Melbourne, Australia
| | - Sylvie Jaillard
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Melbourne, Australia
- INSERM, Institut de Recherche en Santé, Environement et Travail, University of Rennes, 9 Av. du Professeur Léon Bernard, 35000, Rennes, France
- CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, 2 rue Henri Le Guilloux, 35033 Rennes CEDEX 9F-35033, France
| | - Gorjana Robevska
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Melbourne, Australia
| | - Jocelyn A van den Bergen
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Melbourne, Australia
| | - Shirin Shahbazi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran Province, Tehran, Jalal Al Ahmad St, P9CJ+HC9, Iran
| | - Ambro van Hoof
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center at Houston, 7000 Fannin, Suite 1706, Houston, TX 77030, USA
| | - Katie L Ayers
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Grattan Street, Parkville, VIC 3010, Melbourne, Australia
| | - Gary R Hime
- Department of Anatomy and Physiology, The University of Melbourne, Grattan Street, Parkville, VIC 3010, Melbourne, Australia
| | - Andrew H Sinclair
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Grattan Street, Parkville, VIC 3010, Melbourne, Australia
| | - Elena J Tucker
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Grattan Street, Parkville, VIC 3010, Melbourne, Australia
| |
Collapse
|
20
|
Cohen A, Rossetti R, Florsheim N, Samson AO, Renbaum P, Carbone E, Persani L, Levy-Lahad E, Abu-Libdeh A, Zangen D. A Novel Homozygous BMP15 Mutation Causes Ovarian Dysgenesis and Primary Amenorrhea. J Endocr Soc 2025; 9:bvae221. [PMID: 39850788 PMCID: PMC11756294 DOI: 10.1210/jendso/bvae221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Indexed: 01/25/2025] Open
Abstract
Context Despite a growing number of studies, the genetic etiology in many cases of ovarian dysgenesis is incompletely understood. Objectives This work aimed to study the genetic etiology causing absence of spontaneous pubertal development, hypergonadotropic hypogonadism, and primary amenorrhea in 2 sisters. Methods Whole-exome sequencing was performed on DNA extracted from peripheral lymphocytes of 2 Palestinian sisters born to consanguineous parents. Following a BMP15 variant identification, confirming genetic segregation studies were performed in family members. Three-dimensional (3D) modeling for BMP15 dimer and BMP15-GDF-9 heterodimer were followed by functional studies in human ovarian COV434 granulosa cells cotransfected with plasmid harboring either the variant or a wild-type (WT) control, and a second plasmid harboring a luciferase-reporter-gene with a BMP-responsive element. Results A novel homozygous c.G959A/p.C320Y BMP15 mutation was identified in both sisters, and segregated with the disease in the family. By 3D-structure modeling, the mutations were predicted to damage a cysteine-knot motif, disrupt BMP15 dimerization, and severely impair activation of the BMP pathway. The homologous mutation C53Y occurring and identified spontaneously in sheep results in sterility in homozygotes, mimicking the human phenotype here. A 3.8-fold decrease in BMP15 signaling was observed in vitro in cells expressing the homozygous BMP15 mutant when compared to the WT control. Conclusion The novel homozygous missense C320Y mutation is the first homozygous human BMP15 variant causing impaired signaling ability, which correlates with the predicted 3D-structural changes leading to ovarian dysgenesis. The homologous mutation in sheep mimics the human phenotype by infertility. Beyond genetic counseling, and considering ovarian preservation, the ovine model enables further elucidation and interventions in the BMP signaling.
Collapse
Affiliation(s)
- Amitay Cohen
- Division of Pediatric Endocrinology, Hadassah Medical Center, Jerusalem 91240, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Raffaella Rossetti
- Department of Endocrine and Metabolic Diseases, IRCCS Instituto Auxologico Italiano, Milan 20149, Italy
| | - Natan Florsheim
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
- Institute of Medical Genetics, Shaare Zedek Medical Center, Jerusalem 91031, Israel
| | | | - Paul Renbaum
- Institute of Medical Genetics, Shaare Zedek Medical Center, Jerusalem 91031, Israel
| | - Erika Carbone
- Department of Endocrine and Metabolic Diseases, IRCCS Instituto Auxologico Italiano, Milan 20149, Italy
| | - Luca Persani
- Department of Endocrine and Metabolic Diseases, IRCCS Instituto Auxologico Italiano, Milan 20149, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan 20133, Italy
| | - Ephrat Levy-Lahad
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
- Institute of Medical Genetics, Shaare Zedek Medical Center, Jerusalem 91031, Israel
| | - Abdulsalam Abu-Libdeh
- Division of Pediatric Endocrinology, Hadassah Medical Center, Jerusalem 91240, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
- Department of Pediatrics, Makassed Islamic Charitable Hospital, Jerusalem 19482, Israel
| | - David Zangen
- Division of Pediatric Endocrinology, Hadassah Medical Center, Jerusalem 91240, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| |
Collapse
|
21
|
Li D, Liu Y, Hui Y, Li B, Hao C. A Glimpse of Research Trends and Frontiers in the Etiology of Premature Ovarian Insufficiency via Bibliometric Analysis. Endocr Metab Immune Disord Drug Targets 2025; 25:310-325. [PMID: 38919087 DOI: 10.2174/0118715303313887240624071238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024]
Abstract
INTRODUCTION Premature Ovarian Insufficiency (POI) is the most common reproductive aging disorder in women of reproductive age, which is characterized by decreased ovarian function in women before the age of 40. Etiology research of POI has garnered interest and attention from scholars worldwide over the past decades. METHODS However, to the best of our knowledge, no comprehensive survey with bibliometric analysis has been conducted yet on the research trends of POI etiology. This article aimed to analyze current scientific findings on the etiology of POI, offering innovative ideas for further research. Research articles on the etiology of POI from 1994 to 2023 were collected from the Web of Science Core Collection. A total of 456 research articles were included, and the total number of publications increased annually. We used VOSviewer and bibliometric.com to analyze the keywords, terms, institution, publication country/region, author name, publication journal, and the sum of times the articles have been cited. RESULTS This study has shown that a research hotspot is the genetic etiology of POI; however, there is still a lack of research on the impact of epigenetic alterations, iatrogenic injuries, environmental pollution, social stress, and unhealthy lifestyles on the pathogenesis of POI. CONCLUSION The factors illustrated here represent potential future directions for POI etiology research and warrant more attention from researchers.
Collapse
Affiliation(s)
- Duan Li
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, China
- College of Medicine, Qingdao University, Qingdao, China
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Yingxue Liu
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, China
- College of Medicine, Qingdao University, Qingdao, China
| | - Yameng Hui
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Cuifang Hao
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, China
- College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
22
|
Huang Y, Zhang Q, Shen D, Bao X. Mechanisms of He Shi Yu Lin formula in treating premature ovarian insufficiency: insights from network pharmacology and animal experiments. J Ovarian Res 2024; 17:254. [PMID: 39731132 DOI: 10.1186/s13048-024-01575-1] [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: 09/19/2024] [Accepted: 12/08/2024] [Indexed: 12/29/2024] Open
Abstract
OBJECTIVE He Shi Yu Lin Formula (HSYLF) is a clinically proven prescription for treating premature ovarian insufficiency (POI), and has shown a good curative effect. However, its molecular mechanisms are unclear. This study aimed to investigate the molecular mechanisms of HSYLF and clarify how network pharmacology analysis guides the design of animal experiments, including the selection of effective treatment doses and key targets, to ensure the relevance of the experimental results. METHODS Network pharmacology, molecular docking, and animal experiments were utilized to investigate the effects of HSYLF. Key targets were identified by intersecting herb and disease targets to construct protein-protein interaction and "active components-intersection targets-disease" networks. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses were performed using the clusterProfiler package in R. A total of 50 specific pathogen-free female mice of reproductive age were included in the animal experiments. They were divided into five groups: the positive control group, the high-dose HSYLF group, the low-dose HSYLF group, the model blank group, and the normal control group, to evaluate the serum anti-müllerian hormone levels, mitochondrial morphology in oocytes, the levels of reactive oxygen species (ROS), and mitochondrial membrane potential. RESULTS Network pharmacology identified 204 active components connecting 219 key therapeutic targets for POI. Gene Ontology enrichment analysis indicated that the anti-POI targets of HSYLF mainly regulated response to xenobiotic stimulus, cellular response to chemical stress, and response to oxidative stress; and the Kyoto Encyclopedia of Genes and Genomes pathway analysis suggested the primary pathways, including lipid and atherosclerosis, advanced glycation end product-receptor for advanced glycation end product signaling pathway in diabetic complications, bladder cancer, tumor necrosis factor signaling pathway, and interleukin-17 signaling pathway. The low-dose (33 g/kg/d) HSYLF and high-dose (66 g/kg/d) HSYLF groups exhibited a marked elevation in serum anti-müllerian hormone levels (low-dose group: 2657.63 ± 354.82 PG/ml; high-dose group: 2823.73 ± 316.04 PG/ml) and mitochondrial membrane potential compared to the model blank group (P < 0.05 or P < 0.01), along with a significant decline in fluorescence intensity of 2',7'-dichlorofluorescein for the levels of ROS in oocytes (P < 0.05 or P < 0.01). Additionally, both groups showed varying degrees of improvement in the morphology, quantity, and distribution of mitochondria. CONCLUSION This study provides definite evidence for the molecular mechanism by which HSYLF treats POI by decreasing mitochondrial ROS, increasing membrane potential, and improving mitochondrial function. The results from active components of HSYLF and their related key targets also confirmed the characteristics of its multi-component, multi-target, multi-pathway, and overall regulatory effects on POI. Further research regarding the mechanisms is required to generalize these results, and the deeper clinical value of HSYLF also needs to be investigated in the future.
Collapse
Affiliation(s)
- Yun Huang
- TCM Gynecology Department, Hangzhou Hospital of Traditional Chinese Medicine, NO.453 Ti Yuchang Road, Hangzhou, 310007, Zhejiang, China.
- Geriatric Department, Hangzhou Hospital of Traditional Chinese Medicine, NO.453 Ti Yuchang Road, Hangzhou, 310007, Zhejiang, China.
| | - Qin Zhang
- TCM Gynecology Department, Hangzhou Hospital of Traditional Chinese Medicine, NO.453 Ti Yuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Dan Shen
- TCM Gynecology Department, Hangzhou Hospital of Traditional Chinese Medicine, NO.453 Ti Yuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Xi Bao
- TCM Gynecology Department, Hangzhou Hospital of Traditional Chinese Medicine, NO.453 Ti Yuchang Road, Hangzhou, 310007, Zhejiang, China.
| |
Collapse
|
23
|
Qiu Z, Huang EYZ, Li Y, Du J, Kan J. Association of dietary preferences with primary ovarian insufficiency (POI): a mendelian randomization-based analysis. BMC Womens Health 2024; 24:652. [PMID: 39709418 DOI: 10.1186/s12905-024-03488-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/28/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Primary ovarian insufficiency (POI) is a critical cause of infertility and is increasingly recognized as a complex metabolic disorder. Dietary factors may influence the risk of POI, but causal relationships remain unclear. METHODS We conducted an MR study using genetic instrumental variables for 83 dietary preferences from the UK Biobank, with the Inverse Variance Weighted method as the primary analysis. RESULTS Consumption of butter and full-fat dairy products was strongly associated with an increased risk of primary ovarian insufficiency (POI). Women who consumed butter had nearly ten times the risk of developing POI (OR = 9.54, p = 0.048), while full-cream milk was associated with an even greater risk (OR = 29.22, p = 0.018). Interestingly, semi-skimmed milk, despite its lower fat content, also showed a significant positive association with POI (OR > 100, p = 0.008). In contrast, dietary patterns including oily fish and pork were protective against POI. Oily fish, rich in omega-3 fatty acids, was linked to a 82% reduced risk of POI (OR = 0.18, p = 0.008), and pork consumption also showed a protective effect (OR = 0.13, p = 0.041). Additionally, women who did not consume eggs had a significantly lower risk of POI (OR < 0.001, p = 0.044). CONCLUSION This study demonstrates that high-fat dairy products may increase the risk of POI, while oily fish and pork consumption could offer protective effects. These findings providing a foundation for future clinical and public health strategies targeting reproductive health.
Collapse
Affiliation(s)
- Zhengqi Qiu
- The Institute of Mental Psychology, School of Health Management, Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong-Hong Kong-Macao Greater Bay Area Medical and Health Industry High Quality Development Rule of Law Guarantee Research Center, Guangzhou Medical University, 510370, Guangzhou, China
| | | | - Yufei Li
- The Institute of Mental Psychology, School of Health Management, Guangzhou Medical University, Guangzhou, 510370, China
| | - Jun Du
- Nutrilite Health Institute, 720 Cailun Road, 201203, Shanghai, China
| | - Juntao Kan
- Nutrilite Health Institute, 720 Cailun Road, 201203, Shanghai, China.
| |
Collapse
|
24
|
Du Y, Hu Y, Sheng Y, Zhu T, Liu S, Ding H, Guan Y. Primary ovarian insufficiency consequence of autoimmune diseases: a bidirectional two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1417896. [PMID: 39717103 PMCID: PMC11663653 DOI: 10.3389/fendo.2024.1417896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/20/2024] [Indexed: 12/25/2024] Open
Abstract
Background Observational studies suggest the risk of primary ovarian insufficiency (POI) is increased in autoimmune disorders (AIDs), but it is unclear whether there is a causal relationship. Therefore, we aimed to investigate the bidirectional causality between 20 AIDs and POI using Mendelian randomization (MR) analysis. Methods A bidirectional two-sample MR investigation was designed by using publicly accessible summary-level data from genome-wide association studies (GWAS). The inverse variance weighted (IVW) method was performed as the main analysis, supplemented by several sensitivity analyses. Cochran Q test was used to evaluate SNP estimate heterogeneity. MR-Egger and MR-PRESSO methods were utilized to detect horizontal pleiotropy. Results The MR analyses revealed that genetically determined coeliac disease (CeD) (OR = 1.124, 95% CI 1.033-1.224, P = 0.007), vitiligo (OR = 1.092, 95% CI 1.003-1.188; P = 0.042), systemic lupus erythematosus (SLE) (OR = 1.122, 95% CI 1.030-1.223, P = 0.008), and selective immunoglobulin A deficiency (SIgAD) (OR = 0.866, 95% CI: 0.776-0.967, P = 0.011) exhibited significant causal relationships with POI. We also found suggestive evidence of positive effect of Addison's disease (AD) towards POI (OR5e-6 = 1.076, 95% CI 1.002-1.154, P = 0.043). Conclusion This comprehensive MR analysis indicated that SLE, CeD, vitiligo, and AD caused an increased risk of POI, SIgAD was associated with a decreased risk of POI. These insights carry profound clinical implications, particularly emphasizing the early intervention for women with AIDs/POI who wish to preserve their reproductive potential or plan for future pregnancies.
Collapse
Affiliation(s)
- Yongming Du
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yichao Hu
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yuehua Sheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Tianhong Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shenping Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Huiqing Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yutao Guan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
25
|
Panay N, Anderson RA, Bennie A, Cedars M, Davies M, Ee C, Gravholt CH, Kalantaridou S, Kallen A, Kim KQ, Misrahi M, Mousa A, Nappi RE, Rocca WA, Ruan X, Teede H, Vermeulen N, Vogt E, Vincent AJ. Evidence-based guideline: premature ovarian insufficiency . Hum Reprod Open 2024; 2024:hoae065. [PMID: 39660328 PMCID: PMC11631070 DOI: 10.1093/hropen/hoae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Indexed: 12/12/2024] Open
Abstract
STUDY QUESTION How should premature/primary ovarian insufficiency (POI) be diagnosed and managed based on the best available evidence from published literature? SUMMARY ANSWER The current guideline provides 145 recommendations on symptoms, diagnosis, causation, sequelae, and treatment of POI. WHAT IS KNOWN ALREADY Premature ovarian insufficiency (POI) presents a significant challenge to women's health, with far-reaching implications, both physically and emotionally. The potential implications include adverse effects on quality of life; fertility; and bone, cardiovascular, and cognitive health. Although hormone therapy (HT) can mitigate some of these effects, many questions still remain regarding the optimal management of POI. STUDY DESIGN SIZE DURATION The guideline was developed according to the structured methodology for development of ESHRE guidelines. Key questions were determined by a group of experts and informed by a scoping survey of women and health care professionals. Literature searches and assessments were then performed. Papers published up to 30 January 2024 and written in English were included in the guideline. An integrity review was conducted for the randomized controlled trials (RCTs) on POI included in the guideline. PARTICIPANTS/MATERIALS SETTING METHODS Based on the collected evidence, recommendations were formulated and discussed within the guideline development group until consensus was reached. Women with lived experience of POI informed the recommendations in general, and particularly on those on provision of care. A stakeholder review was organized after finalization of the draft. The final version was approved by the guideline development group and the ESHRE Executive Committee. MAIN RESULTS AND THE ROLE OF CHANCE New data indicate a higher prevalence of POI, 3.5%, than was previously thought. This guideline aims to help health care professionals to apply best practice care for women with POI. The recent update of the POI guideline covers 40 clinical questions on diagnosis of the condition, the different sequelae, including bone, cardiovascular, neurological and sexual function, fertility and general well-being, and treatment options, including HT. The list of clinical questions was expanded from the previous iteration of the guideline (2015) based on the scoping survey and appreciation of emerging knowledge of POI. Questions were added on the role of anti-Müllerian hormone (AMH) in the diagnosis of POI, fertility preservation, muscle health, and specific considerations for HT in iatrogenic POI. Additionally, the topic on complementary treatments was extended with specific focus on non-hormonal treatments and lifestyle management options. Significant changes from the previous 2015 guideline include the recommendations that only one elevated FSH >25 IU is required for diagnosis of POI, and guidance that AMH testing, repeat FSH measurement, and/or AMH may be required where there is diagnostic uncertainty. Recommendations were also updated regarding genetic testing, estrogen doses and regimens, use of the combined oral contraceptive and testosterone therapy. Women with lived experience of POI informed the recommendations on provision of care. LIMITATIONS REASONS FOR CAUTION The guideline describes different management options, but it must be acknowledged that for most of these options, supporting evidence is limited for POI. WIDER IMPLICATIONS OF THE FINDINGS The guideline provides health care professionals with clear advice on best practice in POI care, based on the best evidence currently available. In addition, a list of research recommendations is provided to guide further studies in POI. STUDY FUNDING/COMPETING INTERESTS The guideline was developed and funded by ESHRE, American Society for Reproductive Medicine (ASRM), Centre for Research Excellence in Women's Health in Reproduction Life (CRE-WHiRL), and International Menopause Society (IMS), covering expenses associated with the guideline meetings, literature searches, and dissemination of the guideline. The guideline group members did not receive payments. N.P. declared grants from Bayer Pharma (research and consultancy) and NIHR-research POISE; consulting fees from Abbott, Astellas, Bayer, Besins, Lawley, Mithra, Theramex, Viatris; honoraria from Astellas, Bayer, Besins, Gedeon Richter, Theramex, Viatris; support for attending meetings and/or travel from Astellas, Bayer, Theramex, Viatris; President, International Menopause Society, Medical Advisory Committee member, British Menopause Society, Patron Daisy Network. A.J.V. declared grants from Amgen Australia, Australian NHMRC, and Australian MRFF; consulting fees from IQ Fertility; honoraria from the Australasian Menopause Society; participation on a Data Safety Monitoring Board or Advisory Board of Astellas; Board Member of the International Menopause Society (2020 to current) and Past president of the Australasian Menopause Society (2017-2019); R.A.A. declared grants from Roche (Research support, to institution), and participation on a Data Safety Monitoring Board of Bayer. M.C. declared grants from NHI; payments or honoraria from Up-to-Date (as editor/reviewer); Board Member of American Society of Reproductive Medicine, and of American Gynecological and Obstetrical Society. M.D. declared (NIHR-HTA Reference Number: NIHR133461; NIHR-HTA Reference Number: NIHR128757; Action Medical Research and Borne: GN2818) consulting fees from a small personal medical practice, support for attending meetings and/or travel from ESHRE, Bayer and UCLH special Trustees; Participation on the Advisory Board of the British Menopause Society, UKSTORE project, the Progress Educational Trust, and the Turner Syndrome Support Society UK; Leadership or fiduciary roles in the British Fertility Society (Trustee), Elizabeth Garrett Anderson Hospital Charity (chair of Trustees), and the Essex Wynter charitable trust (Trustee). C.E. declared being Chair of a SIG from the Royal Australian College of General Practitioners Integrative Medicine Specific Interest Group and Program Lead for Next Practice Western Sydney Integrative Health. C.H.G. declared grants from Novo Nordisk Foundation (Nos. NNF15OC0016474 and NNF20OC0060610), sygesikringen danmark (No 2022-0189), and the Independent Research Fund Denmark (Nos. 0134-00406 and 0134-00130B); consulting fees from Novo Nordisk, Merck, and Astra Zeneca. S.K. declared grants from Roche diagnostics. A.K. declared grants from NIH R01 5R01HD101475; consulting fees as Medical Reviewer for Flo and for Healthline; honoraria as Medical Consultant for Summus; support for attending meetings from the Reproductive Scientist Development Program; Society for Reproductive Investigation Council Member and Society for Assisted Reproduction Registry/Validation Chair; R.E.N. declared consulting fees from Astellas, Bayer Pharma, Besins Healthcare, Fidia, Theramex; honoraria from Abbott, Astellas, Exeltis, Fidia, Gedeon Richter, Merck & Co, Novo Nordisk, Shionogi Limited, Theramex, Viatris; payment for expert testimony from Vichy Laboratories; Participation in Data Safety Monitoring Board of Advisory board from Astellas and Bayer Healthcare; President elect of the International Menopause Society (IMS). H.T. declared a grant from NHMRC Centre for Research Excellence for women's health in reproductive life. A.B. declared being chair of the Daisy Network Charity. The other authors have no conflicts of interest to declare. DISCLAIMER This guideline represents the views of ESHRE, ASRM, CRE-WHiRL, and IMS, which were achieved after careful consideration of the scientific evidence available at the time of preparation. In the absence of scientific evidence on certain aspects, a consensus between the relevant stakeholders has been obtained. Adherence to these clinical practice guidelines does not guarantee a successful or specific outcome, nor does it establish a standard of care. Clinical practice guidelines do not replace the need for application of clinical judgement to each individual presentation, nor variations based on locality and facility type. The collaborating societies make no warranty, expressed or implied, regarding the clinical practice guidelines and specifically exclude any warranties of merchantability and fitness for a particular use or purpose. (Full disclaimer available at www.eshre.eu/guidelines.).
Collapse
Affiliation(s)
- Nick Panay
- Queen Charlotte’s and Chelsea Hospital, Imperial College London, London, UK
| | - Richard A Anderson
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | | | - Marcelle Cedars
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, School of Medicine, San Francisco, CA, USA
| | - Melanie Davies
- Reproductive Medicine Unit, University College London Hospital, London, UK
| | - Carolyn Ee
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Claus H Gravholt
- Department of Endocrinology, Aarhus University Hospital, Aarhus, Denmark
| | - Sophia Kalantaridou
- Third Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Athens, Greece
| | - Amanda Kallen
- Division of Reproductive Endocrinology and Infertility, University of Vermont Larner College of Medicine, Burlington, VT, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Kimberly Q Kim
- RESOLVE: The National Infertility Association, McLean, VA, USA
| | - Micheline Misrahi
- Faculté de Médecine, Hôpital Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Aya Mousa
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Clayton, Australia
| | - Rossella E Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS San Matteo Foundation, Pavia, Italy
| | - Walter A Rocca
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Beijing, China
| | - Helena Teede
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Clayton, Australia
| | | | - Elinor Vogt
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Amanda J Vincent
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Clayton, Australia
| |
Collapse
|
26
|
Chico-Sordo L, García-Velasco JA. MicroRNAs as Biomarkers and Therapeutic Targets in Female Infertility. Int J Mol Sci 2024; 25:12979. [PMID: 39684688 PMCID: PMC11640832 DOI: 10.3390/ijms252312979] [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: 11/14/2024] [Revised: 12/01/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
The study of microRNAs (miRNAs) has emerged in recent decades as a key approach to understanding the pathophysiology of many diseases, exploring their potential role as biomarkers, and testing their use as future treatments. Not only have neurological, cardiovascular diseases, or cancer benefited from this research but also infertility. Female infertility, as a disease, involves alterations at multiple levels, such as ovarian and uterine alterations. This review compiles the latest studies published in humans that link female disorders that affect fertility with altered miRNA profiles. Studies on ovarian alterations, including diminished ovarian reserve (DOR), poor ovarian response to stimulation (POR), premature ovarian insufficiency (POI), and polycystic ovary syndrome (PCOS), are summarized and classified based on the expression and type of sample analyzed. Regarding uterine disorders, this review highlights upregulated and downregulated miRNAs primarily identified as biomarkers for endometriosis, adenomyosis, decreased endometrial receptivity, and implantation failure. However, despite the large number of studies in this field, the same limitations that reduce reproducibility are often observed. Therefore, at the end of this review, the main limitations of this type of study are described, as well as specific precautions or safety measures that should be considered when handling miRNAs.
Collapse
Affiliation(s)
- Lucía Chico-Sordo
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain;
| | - Juan A. García-Velasco
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain;
- IVIRMA Global Research Alliance, IVIRMA Madrid, 28023 Madrid, Spain
- School of Health Sciences, Medical Specialties and Public Health, Obstetrics and Gynecology Area, Rey Juan Carlos University Alcorcón, 28922 Madrid, Spain
| |
Collapse
|
27
|
Huang S, Zhang D, Shi X, Zhang Y, Wang X, She Y, Liang C, Li X, Zaslawski C. Acupuncture and related therapies for anxiety and depression in patients with premature ovarian insufficiency and diminished ovarian reserve: a systematic review and meta-analysis. Front Psychiatry 2024; 15:1495418. [PMID: 39687777 PMCID: PMC11647530 DOI: 10.3389/fpsyt.2024.1495418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/06/2024] [Indexed: 12/18/2024] Open
Abstract
Background The decreased ovarian function has a negative impact on the mental health of women and increases the risk of anxiety and depression. A growing number of clinical studies have demonstrated that acupuncture-related therapies can effectively and safely restore hormone levels and improve ovarian reserve function. However, the effectiveness of acupuncture-related therapies in alleviating anxiety and depression symptoms in patients with ovarian hypofunction has not been thoroughly evaluated. Therefore, this study conducted a systematic review and meta-analysis to assess the impact of the different acupuncture-related therapies on the mental health of patients with ovarian hypofunction. Methods We comprehensively searched eight famous databases for randomized controlled trials up to October 30, 2024. Databases include PubMed, Web of Science, EMBASE and Cochrane Library, China Biomedical (CBM), China National Knowledge Infrastructure (CNKI), Wanfang Database and VIP Database. Results The study included 12 RCTs, involving 780 patients with ovarian hypofunction, including 403 patients with POI, 297 patients with DOR, and 80 patients with POF. Acupuncture-related therapy was obviously superior to hormone therapy in relieving anxiety symptoms (SMD: -0.90; 95%CI: -1.28, -0.53; P<0.000 01) and depressive symptoms (SMD: -0.82; 95% CI: -1.25, -0.40; P=0.0001). Conclusions Acupuncture-related therapy was more effective than hormone therapy in improving anxiety and depression symptoms in patients with ovarian hypofunction. This study supports the use of acupuncture-related therapies for women experiencing decreased ovarian function associated with mental health issues. Systematic review registration https://www.crd.york.ac.uk/, identifier CRD42023488015.
Collapse
Affiliation(s)
- Sidan Huang
- School of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Danni Zhang
- School of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Xuliang Shi
- School of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- Hebei International Joint Research Center for Dominant Diseases in Chinese Medicine and Acupuncture, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yi Zhang
- School of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Xuesong Wang
- School of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yanfen She
- School of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- Hebei International Joint Research Center for Dominant Diseases in Chinese Medicine and Acupuncture, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Ce Liang
- Pharmacological Lab of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Hebei Traditional Chinese Medicine Formula Granule Technology Innovation Center, Shijiazhuang, Hebei, China
| | - Xinyue Li
- School of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Christopher Zaslawski
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
28
|
Panay N, Anderson RA, Bennie A, Cedars M, Davies M, Ee C, Gravholt CH, Kalantaridou S, Kallen A, Kim KQ, Misrahi M, Mousa A, Nappi RE, Rocca WA, Ruan X, Teede H, Vermeulen N, Vogt E, Vincent AJ. Evidence-based guideline: premature ovarian insufficiency †‡. Climacteric 2024; 27:510-520. [PMID: 39647506 DOI: 10.1080/13697137.2024.2423213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 12/10/2024]
Abstract
STUDY QUESTION How should premature/primary ovarian insufficiency (POI) be diagnosed and managed, based on the best available evidence from published literature? SUMMARY ANSWER The current guideline provides 145 recommendations on symptoms, diagnosis, causation, sequelae and treatment of POI. WHAT IS KNOWN ALREADY POI presents a significant challenge to women's health, with far-reaching implications, both physically and emotionally. The potential implications include adverse effects on quality of life, on fertility and on bone, cardiovascular and cognitive health. Although hormone therapy (HT) can mitigate some of these effects, many questions still remain regarding the optimal management of POI. STUDY DESIGN, SIZE, DURATION The guideline was developed according to the structured methodology for development of European Society of Human Reproduction and Embryology (ESHRE) guidelines. Key questions were determined by a group of experts and informed by a scoping survey of women and healthcare professionals. Literature searches and assessment were then performed. Papers published up to 30 January 2024 and written in English were included in the guideline. An integrity review was conducted for the randomized controlled trials on POI included in the guideline. PARTICIPANTS/MATERIALS, SETTING, METHODS Based on the collected evidence, recommendations were formulated and discussed within the guideline development group until consensus was reached. Women with lived experience of POI informed the recommendations in general, and particularly those on provision of care. A stakeholder review was organized after finalization of the draft. The final version was approved by the guideline development group and the ESHRE Executive Committee. MAIN RESULTS AND THE ROLE OF CHANCE New data indicate a higher prevalence of POI, 3.5%, than was previously thought. This guideline aims to help healthcare professionals apply best practice care for women with POI. The recent update of the POI guideline covers 40 clinical questions on diagnosis of the condition, the different sequelae, including bone, cardiovascular, neurological and sexual function, fertility and general well-being, and treatment options, including HT. The list of clinical questions was expanded from the previous iteration of the guideline (2015) based on the scoping survey and appreciation of emerging knowledge of POI. Questions were added on the role of anti-Müllerian hormone (AMH) in the diagnosis of POI, fertility preservation, muscle health and specific considerations for HT in iatrogenic POI. Additionally, the topic on complementary treatments was extended with specific focus on non-hormonal treatments and lifestyle management options. Significant changes from the previous 2015 guideline include the recommendations that only one elevated follicle stimulating hormone (FSH) >25 IU is required for diagnosis of POI and guidance that AMH testing, repeat FSH measurement and/or AMH may be required where there is diagnostic uncertainty. Recommendations were also updated regarding genetic testing, estrogen doses and regimens, use of the combined oral contraceptive and testosterone therapy. Women with lived experience of POI informed the recommendations on provision of care. LIMITATIONS, REASONS FOR CAUTION The guideline describes different management options, but it must be acknowledged that for most of these options, supporting evidence is limited for POI. WIDER IMPLICATIONS OF THE FINDINGS The guideline provides healthcare professionals with clear advice on best practice in POI care, based on the best evidence currently available. In addition, a list of research recommendations is provided to guide further studies in POI.
Collapse
Affiliation(s)
- Nick Panay
- Queen Charlotte's and Chelsea Hospital, Imperial College London, London, UK
| | - Richard A Anderson
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, UK
| | | | - Marcelle Cedars
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Melanie Davies
- Reproductive Medicine Unit, University College London Hospital, London, UK
| | - Carolyn Ee
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Claus H Gravholt
- Department of Endocrinology, Aarhus University Hospital, Aarhus, Denmark
| | - Sophia Kalantaridou
- Third Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Athens, Greece
| | - Amanda Kallen
- Division of Reproductive Endocrinology and Infertility, University of Vermont Larner College of Medicine, Burlington, VT, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Kimberly Q Kim
- RESOLVE, The National Infertility Association, McLean, VA, USA
| | - Micheline Misrahi
- Université Paris Saclay, Faculté de Médecine, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Aya Mousa
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Clayton, VIC, Australia
| | - Rossella E Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS San Matteo Foundation, Pavia, Italy
| | - Walter A Rocca
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Beijing, China
| | - Helena Teede
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Clayton, VIC, Australia
| | | | - Elinor Vogt
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Amanda J Vincent
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Clayton, VIC, Australia
| |
Collapse
|
29
|
Zhang XY, Wang CJ, Shen HH, Jiang F, Shi JL, Wang WJ, Li MQ. Impaired IL-27 signaling aggravates macrophage senescence and sensitizes premature ovarian insufficiency induction by high-fat diet. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167469. [PMID: 39153664 DOI: 10.1016/j.bbadis.2024.167469] [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: 05/31/2024] [Revised: 07/24/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Premature ovarian insufficiency (POI) critically affects female reproductive health, with obesity being a significant and recognized risk factor. Interleukin-27 (IL-27), known for its role in immune modulation and inflammation, has garnered attention in metabolic syndrome research. Nonetheless, the role of these immunometabolic factors on the initiation of POI remains to be unraveled. Our investigation delves into the influence of impaired IL-27 signaling on POI induction, particularly under the challenge of a high-fat diet (HFD). We analyzed patients' serum profiles and established a correlation of increased serum triglycerides with decreased IL-27 levels in POI cases. Experiments on C57BL/6 mice lacking the IL-27 receptor alpha (Il27ra-/-) revealed that when subjected to HFD, these mice developed hallmark POI symptoms. This includes escalated lipid deposition in both liver and ovarian tissues, increased ovarian macrophages cellular aging, and diminished follicle count, all pointing to compromised ovarian function. These findings unveil a novel pathway wherein impaired IL-27 signaling potentiates the onset of POI in the presence of HFD. Understanding the intricate interplay between IL-27, metabolic alterations, and immune dysregulation sheds light on potential therapeutic avenues for managing POI, offering hope for improved reproductive health outcomes.
Collapse
Affiliation(s)
- Xin-Yan Zhang
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Cheng-Jie Wang
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China; Hospital of Obstetrics and Gynecology, Fudan University, 200080, People's Republic of China
| | - Hui-Hui Shen
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200433, People's Republic of China
| | - Jia-Lu Shi
- Hospital of Obstetrics and Gynecology, Fudan University, 200080, People's Republic of China
| | - Wen-Jun Wang
- Hospital of Obstetrics and Gynecology, Fudan University, 200080, People's Republic of China.
| | - Ming-Qing Li
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China; Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, People's Republic of China.
| |
Collapse
|
30
|
Shi W, Wang D, Xue X, Qiao S, Zhang W, Shi J, Huang C. Epigenomic Landscape of Human Cumulus Cells in Premature Ovarian Insufficiency Using Single-Base Resolution Methylome and Hydroxymethylome. J Cell Mol Med 2024; 28:e70284. [PMID: 39706818 DOI: 10.1111/jcmm.70284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024] Open
Abstract
Premature ovarian insufficiency (POI) has recently been reported to be linked with epigenetic changes. Previous studies have focused on the regulation of individual genes associated with ovarian function through single-gene epigenetic variations; however, there is a deficiency in the comprehensive comprehension of the epigenetic profile for POI. Therefore, we conducted a multi-omics study integrating methylation, hydroxymethylation and transcriptome sequencing analyses in cumulus cells from women with POI and their matched controls. Our data revealed significant global increases in methylation and hydroxymethylation levels in POI patients. We observed a predominance of hypermethylated and hyperhydroxymethylated regions across the genome, with methylation in gene bodies negatively correlating with gene expression, especially in promoter regions. Subsequent experimental validation was performed to confirm the involvement of candidate genes (EGR1, EGR2 and DLX5) in ovarian steroid hormone synthesis. Interestingly, our findings indicate that these epigenetic modifications are associated with genes implicated in POI, ovarian function and the epigenetic age clock. This comprehensive epigenetic profile underscores the potential for identifying novel biomarkers and therapeutic targets for POI by unravelling the complex interplay between DNA epigenetics and ovarian function.
Collapse
Affiliation(s)
- Wenhao Shi
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- The Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Dongyang Wang
- The Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Xia Xue
- The Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Sen Qiao
- The Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Wei Zhang
- Internal Medicine Department, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Juanzi Shi
- The Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| |
Collapse
|
31
|
Monterrosa-Castro Á, Monterrosa-Blanco A, Sánchez-Zarza S. Possible association between subclinical hypothyroidism and age at menopause in Colombian women. Gynecol Endocrinol 2024; 40:2334798. [PMID: 38590105 DOI: 10.1080/09513590.2024.2334798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
OBJECTIVE To evaluate the association between subclinical hypothyroidism with early menopause, premature menopause, and last menstrual bleeding before the natural age of menopause. METHODS This was a cross-sectional study conducted in 643 postmenopausal women aged 40-69 years. Groups were formed according to last menstrual episode: ≥45 [Natural age at menopause], 40-44 and [Early menopause], <40 [Premature menopause], and <45 [last menstrual episode before the natural age of menopause]. The Zulewski scale was applied to identify manifestations related to hypothyroidism and subclinical hypothyroidism, diagnosed with a serum TSH > 4.5 µIU/mL plus T4-free between 0.7 and 1.9 ng/dL. RESULTS It was found that 24.4% had the last menstrual episode before the natural age of menopause, 18.6% had early menopause, and 5.7% had premature menopause. Subclinical hypothyroidism was diagnosed in 4.5% of patients. Among women with subclinical hypothyroidism, there was a higher frequency of early menopause, premature menopause, and last menstrual episode before the natural age of menopause, than in women without subclinical hypothyroidism (p < 0.05). Paresthesia (50%) and dry skin (40.7%) were the most reported hypothyroidism-related manifestations. Early menopause, premature menopause, and last menstrual episode before the natural age of menopause were associated with subclinical hypothyroidism, OR: 3.37 [95% CI: 1.40-8.10], OR: 4.31 [95% CI: 1.24-14.97], and OR: 3.57 [95% CI: 1.57-8.10], respectively. CONCLUSIONS The last menstrual episode before the natural age of menopause, early menopause, and premature menopause were significantly associated with a higher chance of subclinical hypothyroidism.
Collapse
Affiliation(s)
- Álvaro Monterrosa-Castro
- Grupo de investigación Salud de la Mujer, Facultad de Medicina, Universidad de Cartagena, Cartagena, Colombia
| | - Angelica Monterrosa-Blanco
- Grupo de investigación Salud de la Mujer, Facultad de Medicina, Universidad de Cartagena, Cartagena, Colombia
| | - Sandra Sánchez-Zarza
- Grupo de investigación Salud de la Mujer, Facultad de Medicina, Universidad de Cartagena, Cartagena, Colombia
- Instituto de Previsión Social (IPS). Hospital Central, Dr. Emilio Cubas, Facultad de Ciencias de la Salud, Universidad Católica Nuestra Señora de la Asunción', Asunción, Paraguay
| |
Collapse
|
32
|
Zhao X, Yu Z, Wang X, Li X, Liu Y, Wang L. The administration of human amniotic epithelial cells in premature ovarian insufficiency: From preclinical to clinical. Gynecol Endocrinol 2024; 40:2382818. [PMID: 39039858 DOI: 10.1080/09513590.2024.2382818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024] Open
Abstract
Premature ovarian insufficiency (POI) or premature ovarian failure (POF) is a multifactorial disorder occurring in reproductive-age women, characterized by elevated levels of follicle-stimulating hormone (FSH) and irregular or absent menstrual cycles, often accompanied by perimenopausal symptoms and infertility. While assisted reproductive technology can address the reproductive aspirations of some POI-affected women, it is hindered by issues such as exorbitant expenses, substantial risks, and poor rates of conception. Encouragingly, extensive research is exploring novel approaches to enhance fertility, particularly in the realm of stem cell therapy, showcasing both feasibility and significant potential. Human amniotic epithelial cells (hAECs) from discarded placental tissues are crucial in regenerative medicine for their pluripotency, low immunogenicity, non-tumorigenicity, accessibility, and minimal ethical concerns. Preclinical studies highlight the underlying mechanisms and therapeutic effects of hAECs in POI treatment, and current research is focusing on innovative interventions to augment hAECs' efficacy. However, despite these strides, overcoming application challenges is essential for successful clinical translation. This paper conducted a comprehensive analysis of the aforementioned issues, examining the prospects and challenges of hAECs in POI, with the aim of providing some insights for future research and clinical practice.
Collapse
Affiliation(s)
- Xiaojing Zhao
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhongna Yu
- Department of Gynecology, The Affiliated People's Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Xinrun Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaojing Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yang Liu
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liang Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
33
|
Jamali Z, Khalili P, Ayoobi F, Vatankhah H, Esmaeili-Nadimi A, Ranjbar FE, Vatanparast M. Type of menopause, age of menopause and cardiovascular disease: a cross-sectional study based on data from Rafsanjan cohort study. BMC Womens Health 2024; 24:626. [PMID: 39593060 PMCID: PMC11590513 DOI: 10.1186/s12905-024-03452-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: 04/09/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Cardiovascular disease is the leading cause of death among women, but sex-specific risk factors are incompletely understood. In this study, we aimed to assess the associations between the type of menopause, and age at natural menopause with the odds of cardiovascular disease (CVD), and coronary heart disease (CHD). METHODS This cross-sectional study is a part of data from the Rafsanjan Cohort Study (RCS) which is a branch of the Prospective Epidemiological Research Studies in Iran (PERSIAN). A sample of 1767 postmenopausal women were included. The diagnosis for CVD and CHD was based on self-report questionnaires. Menopause age was categorized as < 40, 40-44, 45-49, and ≥ 50. Also, the menopause types were classified as natural and induced menopause (surgery or chemotherapy). The association was evaluated by logistic regressions. RESULTS The menopause age < 40 years had higher odds of CVD compared to women with menopause age > 40 years (OR: 2.66; 95%CI 1.29-5.48). Women with induced menopause had higher odds of CVD compared to women with natural menopause (OR = 1.44, 95% CI 1.04-1.98). In terms of the odds of CHD, the results showed that the odds of CHD increased in menopause age < 40 years and induced menopause compared to reference groups (OR: 2.49, 95% CI 1.15-5.37, OR = 1.48; 95% CI 1.06-2.07, respectively). CONCLUSION Premature menopause and induced menopause should be considered as important risk factors for CVD, and CHD. Health policymakers should pay more attention to the type of menopause and the age of menopause in postmenopausal women to predict the risk of CVD and preventive strategies.
Collapse
Affiliation(s)
- Zahra Jamali
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Clinical Research Development Unit (CRDU), Niknafs Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Parvin Khalili
- Social Determinants of Health Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Epidemiology, School of Public Health, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Ayoobi
- Occupational Safety and Health Research Center, NICICO, World Safety Organization and Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hajar Vatankhah
- Clinical Research Development Unit (CRDU), Niknafs Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Obstetrics and Gynecology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Esmaeili-Nadimi
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Cardiology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Faezeh Esmaeili Ranjbar
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mahboubeh Vatanparast
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
34
|
Tang S, Guo T, Song C, Wang L, Zhang J, Rajkovic A, Lin X, Chen S, Liu Y, Tian W, Wu B, Wang S, Wang W, Lai Y, Wang A, Xu S, Jin L, Ke H, Zhao S, Li Y, Qin Y, Zhang F, Chen ZJ. MGA loss-of-function variants cause premature ovarian insufficiency. J Clin Invest 2024; 134:e183758. [PMID: 39545409 PMCID: PMC11563689 DOI: 10.1172/jci183758] [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: 06/11/2024] [Accepted: 09/20/2024] [Indexed: 11/17/2024] Open
Abstract
Although premature ovarian insufficiency (POI), a common cause of female infertility and subfertility, has a well-established hereditary component, the genetic factors currently implicated in POI account for only a limited proportion of cases. Here, using an exome-wide, gene-based case-control analysis in a discovery cohort comprising 1,027 POI cases and 2,733 ethnically matched women controls from China, we found that heterozygous loss-of-function (LoF) variants of MAX dimerization protein (MGA) were significantly enriched in the discovery cohort, accounting for 2.6% of POI cases, while no MGA LoF variants were found in the matched control females. Further exome screening was conducted in 4 additional POI cohorts (2 from China and 2 from the United States) for replication studies, and we identified heterozygous MGA LoF variants in 1.0%, 1.4%, 1.0%, and 1.0% of POI cases, respectively. Overall, a total of 37 distinct heterozygous MGA LoF variants were discovered in 38 POI cases, accounting for approximately 2.0% of the total 1,910 POI cases analyzed in this study. Accordingly, Mga+/- female mice were subfertile, exhibiting shorter reproductive lifespan and decreased follicle number compared with WT, mimicking the observed phenotype in humans. Our findings highlight the essential role of MGA deficiency for impaired female reproductive ability.
Collapse
Affiliation(s)
- Shuyan Tang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
| | - Ting Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Chengcheng Song
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
| | - Lingbo Wang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Jun Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Aleksandar Rajkovic
- Department of Pathology, Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| | - Xiaoqi Lin
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
| | - Shiling Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yujun Liu
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
| | - Weidong Tian
- School of Life Sciences, Fudan University, Shanghai, China
| | - Bangguo Wu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenwen Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunhui Lai
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ao Wang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuhua Xu
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
- School of Life Sciences, Fudan University, Shanghai, China
| | - Li Jin
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
- School of Life Sciences, Fudan University, Shanghai, China
| | - Hanni Ke
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Shidou Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingying Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
- Shanghai Key Laboratory of Embryo Original Diseases, Soong Ching Ling Institute of Maternity and Child Health, International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (no. 2021RU001), Jinan, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
35
|
Li Y, Wang L, Liu J, Nie G, Yang H. Huyang Yangkun formula regulates the mitochondria pathway of ovarian granulosa cell apoptosis through FTO/m6A-P53 pathway. Front Pharmacol 2024; 15:1491546. [PMID: 39584135 PMCID: PMC11581872 DOI: 10.3389/fphar.2024.1491546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/25/2024] [Indexed: 11/26/2024] Open
Abstract
Background Premature ovarian insufficiency (POI) presents a significant challenge to female reproductive health. The Huyang Yangkun Formula (HYF), a traditional Chinese medicinal formulation, has been utilized in clinical settings for the treatment of POI for over a decade. Nevertheless, the therapeutic application of HYF is considerably constrained by the lack of clarity regarding its underlying mechanism of action. Methods The experimental procedures entailed administering VCD to female Sprague-Dawley rats at a dosage of 160 mg/kg/day over a period of 15 days, succeeded by a 100-day treatment with HYF. Blood serum samples were collected and analyzed using ELISA to quantify the concentrations of Anti-Müllerian Hormone (AMH), Follicle-Stimulating Hormone (FSH), and Estradiol (E2). The levels of N6-methyladenosine (m6A) were assessed through Dot blot analysis and liquid chromatography-tandem mass spectrometry (LC-MS/MS). Western blotting was employed to validate the differential expression of m6A-related catalytic enzymes and apoptosis-related regulators, including BCL-2, BCL-XL, and MCL-1, which may be implicated in the effects of HYF. Certain shRNA-COV434 cell line was constructed for the exploration of molecular mechanism, and then the potential targets were finally verified by MeRIP-qPCR. Results HYF has been identified as having a significant influence on the development of residual ovarian follicles in rats with POI, especially during the initial stages. It was observed that HYF facilitates the progression of escaping antral follicles to full maturation. Additionally, HYF exhibited the capacity to enhance the proliferation of COV434, a human ovarian granulosa cell line, while concurrently inhibiting apoptosis within these cells. Notably, HYF treatment resulted in the downregulation of apoptotic proteins, including BCL-XL, cleaved-caspase 9, cleaved-caspase 3, and Bcl-2. Concurrently, m6A modification is implicated in the regulation of HYF. Both in vitro and in vivo studies indicate that FTO may play a role in the anti-apoptotic mechanisms mediated by m6A in ovarian granulosa cells influenced by HYF. Moreover, employing qPCR and MeRIP-qPCR techniques, P53 has been identified as the target gene for m6A modification mediated by FTO. Conclusion These findings suggest that HYF holds promise as a potential treatment for POI and provide a more comprehensive understanding of the mechanism by which HYF operates, specifically its ability to prevent the BCL-2 mitochondrial apoptosis pathway mediated by P53 in ovarian granulosa cells of POI rats by regulating FTO/m6A-Tp53.
Collapse
Affiliation(s)
- Yang Li
- Department of Gynaecology, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingdi Wang
- Department of Gynecology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jian Liu
- Department of Gynaecology, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangning Nie
- Department of Gynaecology, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongyan Yang
- Department of Gynaecology, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| |
Collapse
|
36
|
Lafraoui I, Heddar A, Cantalloube A, Braham I, Peigné M, Beneteau C, Gricourt S, Poirsier C, Legrand S, Stoeva R, Metayer-Amelot L, Lobersztajn A, Lebrun S, Gruchy N, Abdennebi I, Cedrin-Durnerin I, Fernandez H, Luton D, Torre A, Zagdoun L, Chevalier N, Khrouf M, Mahmoud K, Epelboin S, Catteau-Jonard S, Misrahi M. Genetic Landscape of a Cohort of 120 Patients with Diminished Ovarian Reserve: Correlation with Infertility. Int J Mol Sci 2024; 25:11915. [PMID: 39595984 PMCID: PMC11593603 DOI: 10.3390/ijms252211915] [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: 09/23/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Diminished ovarian reserve (DOR) and primary ovarian insufficiency (POI) are major causes of female infertility. We recently found a monogenic etiology in 29.3% of POI, leading to personalized medicine. The genetic landscape of DOR is unknown. A prospective study (2018-2023) of an international cohort of 120 patients with unexplained DOR was performed using a large custom targeted next-generation sequencing panel including all known POI-causing genes. The diagnostic yield, based on the American College of Medical Genetics, was 24, 2%. Genes belong to different pathways: metabolism and mitochondria (29.7%), follicular growth (24.3%), DNA repair/meiosis (18.9%), aging (16.2%), ovarian development (8.1%), and autophagy (2.7%). Five genes were recurrently found: LMNA, ERCC6, SOX8, POLG, and BMPR1B. Six genes identified in single families with POI were involved in DOR, GNAS, TGFBR3, XPNPEP2, EXO1, BNC1, ATG, highlighting their role in maintaining ovarian reserve. In our cohort, 26 pregnancies were recorded, but no pregnancy was observed when meiosis/DNA repair genes were involved, suggesting severely impaired oocyte quality. Additional studies should confirm these preliminary results. This study with a large NGS panel defines the genetic landscape of a large cohort of DOR. It supports routine genetic diagnosis. Genetics could be a biomarker predicting infertility and progression to POI.
Collapse
Affiliation(s)
- Imène Lafraoui
- Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Faculté de Médecine Paris Saclay, INSERM U1193, 94275 Le Kremlin-Bicêtre, France; (I.L.); (A.H.)
- Laboratoire de Biologie Moléculaire National de Référence-LBMR Pour les Infertilités Génétiques Chez la Femme et l’Homme, Hôpitaux Universitaires Paris Saclay, 94275 Le Kremlin Bicêtre, France
| | - Abdelkader Heddar
- Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Faculté de Médecine Paris Saclay, INSERM U1193, 94275 Le Kremlin-Bicêtre, France; (I.L.); (A.H.)
- Laboratoire de Biologie Moléculaire National de Référence-LBMR Pour les Infertilités Génétiques Chez la Femme et l’Homme, Hôpitaux Universitaires Paris Saclay, 94275 Le Kremlin Bicêtre, France
| | - Adèle Cantalloube
- Service de Gynécologie-Obstétrique, Hôpital Tenon, Hôpitaux Universitaires Paris Centre, 75014 Paris, France; (A.C.); (S.G.); (S.E.)
| | - Inès Braham
- Service d’Endocrinologie, Diabétologie et Médecine de la Reproduction, CHU de Nice, 06000 Nice, France; (I.B.); (N.C.)
| | - Maëliss Peigné
- Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Jean-Verdier, Université Sorbonne Paris Nord, 93430 Bondy, France; (M.P.); (I.C.-D.)
| | - Claire Beneteau
- Service de Génétique Médicale, CHU de Nantes, 44000 Nantes, France;
| | - Solenne Gricourt
- Service de Gynécologie-Obstétrique, Hôpital Tenon, Hôpitaux Universitaires Paris Centre, 75014 Paris, France; (A.C.); (S.G.); (S.E.)
| | - Claire Poirsier
- Departement de Genetique, Centre Hospitalier Universitaire de Reims, 51092 Reims, France;
| | - Stéphanie Legrand
- Centre de Fertilité, Clinique de l’Atlantique, 17138 La Rochelle, France;
| | - Radka Stoeva
- Laboratoire de Génétique Médicale et Cytogénétique, CH Le Mans, 72037 Le Mans, France;
| | - Laure Metayer-Amelot
- Service d’Endocrinologie et Médecine de la Reproduction, CH Le Mans, 72037 Le Mans, France;
| | - Annina Lobersztajn
- Centre de la Fertilité—Paris Est, Nogent sur Marne, 94130 Nogent-sur-Marne, France;
| | - Soizic Lebrun
- Service de Génétique, FHU GenOMedS, CHRU de Tours, 37000 Tours, France;
| | - Nicolas Gruchy
- EA 7450 BioTARGen, FHU G4 Genomics, Service de Génétique Clinique, Departement de Genetique, CHU Côte de Nacre, Université de Caen Normandie UNICAEN, 14000 Caen, France;
| | - Inès Abdennebi
- Centre d’Aide Médicale à la Procréation, Centre Hospitalier Intercommunal de Créteil, 94000 Créteil, France;
| | - Isabelle Cedrin-Durnerin
- Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Jean-Verdier, Université Sorbonne Paris Nord, 93430 Bondy, France; (M.P.); (I.C.-D.)
| | - Hervé Fernandez
- Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicêtre, France; (H.F.); (D.L.)
| | - Dominique Luton
- Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicêtre, France; (H.F.); (D.L.)
| | - Antoine Torre
- Centre d’Assistance Médicale à la Procréation Clinico-Biologique, Centre Hospitalier Sud Francilien Corbeil-Essonnes, 91100 Corbeil-Essonnes, France;
| | - Léonore Zagdoun
- Service de Diabétologie et Endocrinologie, Centre Hospitalier de Mont de Marsan et Pays des Sources, 40024 Mont de Marsan, France;
| | - Nicolas Chevalier
- Service d’Endocrinologie, Diabétologie et Médecine de la Reproduction, CHU de Nice, 06000 Nice, France; (I.B.); (N.C.)
| | - Mohamed Khrouf
- Centre FERTILLIA de Médecine de la Reproduction-Clinique la Rose, Tunis 1053, Tunisia; (M.K.); (K.M.)
| | - Khaled Mahmoud
- Centre FERTILLIA de Médecine de la Reproduction-Clinique la Rose, Tunis 1053, Tunisia; (M.K.); (K.M.)
| | - Sylvie Epelboin
- Service de Gynécologie-Obstétrique, Hôpital Tenon, Hôpitaux Universitaires Paris Centre, 75014 Paris, France; (A.C.); (S.G.); (S.E.)
| | - Sophie Catteau-Jonard
- Service de Gynécologie Endocrinienne, CHU de Lille, Hôpital Jeanne-de-Flandre, 59000 Lille, France;
| | - Micheline Misrahi
- Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Faculté de Médecine Paris Saclay, INSERM U1193, 94275 Le Kremlin-Bicêtre, France; (I.L.); (A.H.)
- Laboratoire de Biologie Moléculaire National de Référence-LBMR Pour les Infertilités Génétiques Chez la Femme et l’Homme, Hôpitaux Universitaires Paris Saclay, 94275 Le Kremlin Bicêtre, France
| |
Collapse
|
37
|
Jayasena CN, Devine K, Barber K, Comninos AN, Conway GS, Crown A, Davies MC, Ewart A, Seal LJ, Smyth A, Turner HE, Webber L, Anderson RA, Quinton R. Society for endocrinology guideline for understanding, diagnosing and treating female hypogonadism. Clin Endocrinol (Oxf) 2024; 101:409-442. [PMID: 39031660 DOI: 10.1111/cen.15097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/18/2024] [Accepted: 05/27/2024] [Indexed: 07/22/2024]
Abstract
Female hypogonadism (FH) is a relatively common endocrine disorder in women of premenopausal age, but there are significant uncertainties and wide variation in its management. Most current guidelines are monospecialty and only address premature ovarian insufficiency (POI); some allude to management in very brief and general terms, and most rely upon the extrapolation of evidence from the studies relating to physiological estrogen deficiency in postmenopausal women. The Society for Endocrinology commissioned new guidance to provide all care providers with a multidisciplinary perspective on managing patients with all forms of FH. It has been compiled using expertise from Endocrinology, Primary Care, Gynaecology and Reproductive Health practices, with contributions from expert patients and a patient support group, to help clinicians best manage FH resulting from both POI and hypothalamo-pituitary disorders, whether organic or functional.
Collapse
Affiliation(s)
- Channa N Jayasena
- Section of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Kerri Devine
- Department of Endocrinology, Diabetes & Metabolism, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK
- Translational & Clinical Research Institute, University of Newcastle-upon-Tyne, Newcastle-upon-Tyne, UK
| | - Katie Barber
- Community Gynaecology (NHS), Principal Medical Limited, Bicester, Oxfordshire, UK
- Oxford Menopause Ltd, Ardington, Wantage, UK
| | - Alexander N Comninos
- Division of Diabetes, Endocrinology & Metabolism, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Gerard S Conway
- Reproductive Medicine Unit, University College London Hospitals, London, UK
| | - Anna Crown
- Department of Endocrinology, Royal Sussex County Hospital, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Melanie C Davies
- Reproductive Medicine Unit, University College London Hospitals, London, UK
| | - Ann Ewart
- Kallman Syndrome and Congenital Hypogonadotropic Hypogonadism Support Group, Dallas, Texas, United States
| | - Leighton J Seal
- Department of Endocrinology, St George's Hospital Medical School, London, UK
| | - Arlene Smyth
- UK Turner Syndrome Support Society, Clydebank, UK
| | - Helen E Turner
- Department of Endocrinology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Lisa Webber
- Department of Obstetrics & Gynaecology, Singapore General Hospital, Singapore
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Richard Quinton
- Section of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK
- Department of Endocrinology, Diabetes & Metabolism, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK
- Translational & Clinical Research Institute, University of Newcastle-upon-Tyne, Newcastle-upon-Tyne, UK
| |
Collapse
|
38
|
Du H, Zeng P, Liu X, Zhang J, Huang Z. Identifying therapeutic targets for primary ovarian insufficiency through integrated genomic analyses. J Ovarian Res 2024; 17:193. [PMID: 39358799 PMCID: PMC11446024 DOI: 10.1186/s13048-024-01524-y] [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: 08/07/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Primary ovarian insufficiency (POI) is a disorder characterized by the premature decline in ovarian function, leading to significant fertility and health impacts on women under 40. The unclear etiology of POI hinders the development of effective treatments, highlighting the need for novel therapeutic targets. METHODS This study employed genome-wide association analysis (GWAS) integrated with expression quantitative trait loci (eQTL) data from the GTEx and eQTLGen databases. Mendelian randomization (MR) and colocalization analyses were conducted to investigate causal relationships between genetic variants and POI and to identify potential therapeutic targets. RESULTS We identified 431 genes with available index cis-eQTL signals, of which four (HM13, FANCE, RAB2A, and MLLT10) were significantly associated with POI. Colocalization analysis revealed strong evidence for FANCE and RAB2A, indicating their potential as therapeutic targets. Subsequent druggability assessments identified FANCE and RAB2A as promising candidates for POI treatment, supported by their involvement in DNA repair and autophagy regulation, respectively. CONCLUSIONS Our study establishes a causal link between specific genes and POI, highlighting FANCE and RAB2A as potential drug targets. These findings provide a foundation for future research and therapeutic development, aiming to improve outcomes for women with POI. Validation in further trials is necessary to confirm these potential targets.
Collapse
Affiliation(s)
- Haihong Du
- Department of Gynecology, Meishan Women and Children's Hospital, Meishan, Sichuan, China
| | - Pengfei Zeng
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xuyi Liu
- Department of Gynecology, Meishan Women and Children's Hospital, Meishan, Sichuan, China
| | - Jun Zhang
- Department of Traditional Chinese Medicine, Meishan Women and Children's Hospital, Meishan, Sichuan, China
| | - Zhonglu Huang
- Department of Gynecology, Meishan Women and Children's Hospital, Meishan, Sichuan, China.
| |
Collapse
|
39
|
La Marca A, Diamanti M. Factors affecting age at menopause and their relationship with ovarian reserve: a comprehensive review. EUR J CONTRACEP REPR 2024; 29:245-255. [PMID: 39007753 DOI: 10.1080/13625187.2024.2375281] [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: 03/26/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024]
Abstract
OBJECTIVE The aim of this article was to discuss all the factors affecting the age at menopause and their correlation with ovarian reserve. MATERIALS AND METHODS A narrative review of original articles was performed using PubMed until December 2023. The following keywords were used to generate the list of citations: 'menopause', 'ovarian reserve' 'oocytes quality and quantity', 'ovarian ageing'. RESULTS Menopause is the final step in the process of ovarian ageing and is influenced by the oocyte pool at birth. Conditions that accelerate follicle depletion during the reproductive lifespan lead to premature ovarian insufficiency (POI) and premature ovarian failure (POF), while a higher ovarian reserve is associated with a delayed time to menopause. Reproductive history, sociodemographic, lifestyle and iatrogenic factors may impact ovarian reserve and the age at menopause. CONCLUSIONS Some factors affecting the age at menopause are modifiable and the risks of early menopause may be preventable. We hypothesise that by addressing these modifiable factors we may also preserve ovarian reserve. However, further interventional studies are needed to evaluate the effects of the described strategies on ovarian reserve.
Collapse
Affiliation(s)
- Antonio La Marca
- Obstetrics and Gynecology Unit, Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Modena, Italy
| | - Marialaura Diamanti
- Obstetrics and Gynecology Unit, Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
40
|
Namavari N, Jokar M, Ghodsian A, Jahromi HK, Rahmanian V. Menopausal state and rheumatoid arthritis: a systematic review and meta-analysis. BMC Rheumatol 2024; 8:48. [PMID: 39350181 PMCID: PMC11441135 DOI: 10.1186/s41927-024-00418-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 09/11/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory condition primarily affecting the joints. The higher prevalence of RA among females, combined with the known effects of sex hormones on immune function, has led researchers to investigate the potential relationship between menopausal status and the risk, severity, or progression of RA. This systematic review and meta-analysis aimed to determine the association between menopause and rheumatoid arthritis. METHODS In 2023, we conducted a comprehensive search across multiple databases, including Google Scholar, Scopus, PubMed/MEDLINE, Science Direct, Web of Science, EMBASE, Springer, and ProQuest. The search aimed to identify studies exploring the association between menopause and rheumatoid arthritis. RESULTS Our analysis revealed that post-menopausal women had a higher risk of developing rheumatoid arthritis compared to pre-menopausal women, with an estimated odds ratio of 1.35 (95% CI: 1.04-1.67). Additionally, women who experienced early menopause (defined as onset before age 45) showed significantly higher odds of developing RA, with an odds ratio of 2.97 (95% CI: 1.73-4.22). CONCLUSION These findings highlight the importance of considering menopausal status when assessing the risk of RA development in women. The results suggest that post-menopausal women, particularly those who experience early menopause, may be at higher risk for developing RA. Further research in this area could provide valuable insights into potential preventive measures and targeted interventions for high-risk individuals.
Collapse
Affiliation(s)
- Negin Namavari
- School of Medicine, Peymaniye Hospital, Jahrom University of Medical Science, Jahrom, Iran
| | - Mohammad Jokar
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada, University of Calgary, Calgary, Canada
| | - Arnoosh Ghodsian
- School of Medicine, Jahrom University of Medical Science, Jahrom, Iran
| | - Hossein Kargar Jahromi
- Research Center for non- Communicable Disease, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Vahid Rahmanian
- Department of Public Health, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran.
| |
Collapse
|
41
|
Federici S, Rossetti R, Moleri S, Munari EV, Frixou M, Bonomi M, Persani L. Primary ovarian insufficiency: update on clinical and genetic findings. Front Endocrinol (Lausanne) 2024; 15:1464803. [PMID: 39391877 PMCID: PMC11466302 DOI: 10.3389/fendo.2024.1464803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/02/2024] [Indexed: 10/12/2024] Open
Abstract
Primary ovarian insufficiency (POI) is a disorder of insufficient ovarian follicle function before the age of 40 years with an estimated prevalence of 3.7% worldwide. Its relevance is emerging due to the increasing number of women desiring conception late or beyond the third decade of their lives. POI clinical presentation is extremely heterogeneous with a possible exordium as primary amenorrhea due to ovarian dysgenesis or with a secondary amenorrhea due to different congenital or acquired abnormalities. POI significantly impacts non only on the fertility prospect of the affected women but also on their general, psychological, sexual quality of life, and, furthermore, on their long-term bone, cardiovascular, and cognitive health. In several cases the underlying cause of POI remains unknown and, thus, these forms are still classified as idiopathic. However, we now know the age of menopause is an inheritable trait and POI has a strong genetic background. This is confirmed by the existence of several candidate genes, experimental and natural models. The most common genetic contributors to POI are the X chromosome-linked defects. Moreover, the variable expressivity of POI defect suggests it can be considered as a multifactorial or oligogenic defect. Here, we present an updated review on clinical findings and on the principal X-linked and autosomal genes involved in syndromic and non-syndromic forms of POI. We also provide current information on the management of the premature hypoestrogenic state as well as on fertility preservation in subjects at risk of POI.
Collapse
Affiliation(s)
- Silvia Federici
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Raffaella Rossetti
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Silvia Moleri
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Elisabetta V. Munari
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Maria Frixou
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Marco Bonomi
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Luca Persani
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| |
Collapse
|
42
|
Li T, Wei Y, Jiao B, Hao R, Zhou B, Bian X, Wang P, Zhou Y, Sun X, Zhang J. Bushen Huoxue formula attenuates lipid accumulation evoking excessive autophagy in premature ovarian insufficiency rats and palmitic acid-challenged KGN cells by modulating lipid metabolism. Front Pharmacol 2024; 15:1425844. [PMID: 39351088 PMCID: PMC11439644 DOI: 10.3389/fphar.2024.1425844] [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: 04/30/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Premature ovarian insufficiency (POI) has affected about 3.7% of women of reproductive age and is a major factor contributing to infertility. Bushen Huoxue formula (BHF), a traditional Chinese medicine prescription, is clinically used to treat POI in China. This study aims to investigate the potential mechanisms of BHF in combating POI using corticosterone-induced rats and palmitic acid (PA)-challenged human ovarian granulosa cells (GCs). Methods Initially, ultra performance liquid chromatography tandem mass spectrometry was employed to analyze the components of BHF. The pharmacodynamic parameters evaluated included body weight, ovaries index, and serum hormone in rats. Follicle numbers were observed using H&E staining. Additionally, PCNA and TUNEL staining were used to assess GCs proliferation and apoptosis, respectively. Lipid accumulation and ROS levels were examined using Oil Red O and ROS staining. Protein expressions were determined by western blot. To probe mechanisms, cell viability and E2 levels in BHF-treated, PA-stimulated GCs were determined using MTT and ELISA, respectively. Cell apoptosis and ROS levels were assessed using TUNEL and ROS staining. Proteins related to lipid metabolism and autophagy in PA-stimulated GCs were studied using agonists. Results Our results shown that BHF effectively normalized serum hormone levels, including follicle-stimulating hormone (FSH), anti-Müllerian hormone (AMH), estradiol (E2), and luteinizing hormone (LH). Concurrently, BHF also significantly reduced follicular atresia and promoted cell proliferation while inhibiting apoptosis in POI rats. Furthermore, BHF mitigated ovarian lipid accumulation by modulating lipid metabolism, which included reducing lipid synthesis (expression of peroxisome proliferator-activated receptor γ and CCAAT/enhancer binding protein α), increasing lipid catabolism (expression of adipose triglyceride lipase), and enhancing lipid oxidation (expression of carnitine palmitoyl transferase 1A). Mechanistically, the therapeutic effects of BHF on POI were linked with alleviation of lipid deposition-induced reactive oxygen species (ROS) accumulation and excessive autophagy, corroborating the results in PA-challenged GCs. After treatment with elesclomol (a ROS inducer) and rapamycin (an autophagy inducer) in GCs, the effects of BHF were almost counteracted under model conditions. Conclusion These findings suggest that BHF alleviates the symptoms of POI by altering lipid metabolism and reducing lipid accumulation-induced ROS and autophagy, offering evidence for BHF's efficacy in treating POI clinically.
Collapse
Affiliation(s)
- Tian Li
- Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
- Clinical College of Traditional Chinese Medicine Hospital in Lishui, Jiangsu Health Vocational College, Nanjing, China
| | - Yao Wei
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Beibie Jiao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Tonglu Hospital of Traditional Chinese Medicine, Tonglu, China
| | - Rui Hao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Beibei Zhou
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinlan Bian
- Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
- Clinical College of Traditional Chinese Medicine Hospital in Lishui, Jiangsu Health Vocational College, Nanjing, China
| | - Peijuan Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yahong Zhou
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Xia Sun
- Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
- Clinical College of Traditional Chinese Medicine Hospital in Lishui, Jiangsu Health Vocational College, Nanjing, China
| | - Jian Zhang
- Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
- Clinical College of Traditional Chinese Medicine Hospital in Lishui, Jiangsu Health Vocational College, Nanjing, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
43
|
Touraine P, Chabbert-Buffet N, Plu-Bureau G, Duranteau L, Sinclair AH, Tucker EJ. Premature ovarian insufficiency. Nat Rev Dis Primers 2024; 10:63. [PMID: 39266563 DOI: 10.1038/s41572-024-00547-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 09/14/2024]
Abstract
Premature ovarian insufficiency (POI) is a cause of infertility and endocrine dysfunction in women, defined by loss of normal, predictable ovarian activity before the age of 40 years. POI is clinically characterized by amenorrhoea (primary or secondary) with raised circulating levels of follicle-stimulating hormone. This condition can occur due to medical interventions such as ovarian surgery or cytotoxic cancer therapy, metabolic and lysosomal storage diseases, infections, chromosomal anomalies and autoimmune diseases. At least 1 in 100 women is affected by POI, including 1 in 1,000 before the age of 30 years. Substantial evidence suggests a genetic basis to POI. However, the cause of idiopathic POI remains unknown in most patients, indicating that gene variants associated with this condition remain to be discovered. Over the past 10 years, tremendous progress has been made in our knowledge of genes involved in POI. Genetic approaches in diagnosis are important as they enable patients with familial POI to be identified, with the opportunity for oocyte preservation. Moreover, genetic approaches could provide a better understanding of disease mechanisms, which will ultimately aid the development of improved treatments.
Collapse
Affiliation(s)
- Philippe Touraine
- Department of Endocrinology and Reproductive Medicine, AP-HP Pitié Salpêtrière Hospital, Sorbonne Université Médecine, Paris, France.
- Inserm U1151 INEM, Necker Hospital, Paris, France.
| | - Nathalie Chabbert-Buffet
- Department of Obstetrics, Gynecology and Reproductive Medicine, Tenon Hospital, AP-HP Sorbonne Université, Paris, France
- INSERM UMR S 938, CDR St Antoine, Paris, France
| | - Genevieve Plu-Bureau
- Department of Medical Gynecology, AP-HP Port Royal-Cochin Hospital, Université Paris Cité, Paris, France
- U1151 EPOPEE Team, Paris, France
| | - Lise Duranteau
- Department of Medical Gynecology, Bicêtre Hospital, AP-HP Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Andrew H Sinclair
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Elena J Tucker
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
44
|
Luo Y, Chen J, Ning J, Sun Y, Chai Y, Xiao F, Huang B, Li G, Tian F, Hao J, Zhang Q, Zhao J, Li Y, Li H. Stem cell-derived extracellular vesicles in premature ovarian failure: an up-to-date meta-analysis of animal studies. J Ovarian Res 2024; 17:182. [PMID: 39252114 PMCID: PMC11382489 DOI: 10.1186/s13048-024-01489-y] [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: 05/14/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND There has been a significant surge in animal studies of stem cell-derived extracellular vesicles (EVs) therapy for the treatment of premature ovarian failure (POF) but its efficacy remains unknown and a comprehensive and up-to-date meta-analysis is lacking. Before clinical translation, it is crucial to thoroughly understand the overall impact of stem cell-derived EVs on POF. METHODS PubMed, EMBASE, Cochrane Library, Web of Science were searched up to February 18, 2024. The risk of bias was evaluated according to Cochrane Handbook criteria, while quality of evidence was assessed using the SYRCLE system. The PRISMA guidance was followed. Trial sequential analysis was conducted to assess outcomes, and sensitivity analysis and publication bias analysis were performed using Stata 14. RESULTS Data from 25 studies involving 339 animals were extracted and analyzed. The analysis revealed significant findings: stem cell-derived EVs increase ovary weight (SMD = 3.88; 95% CI: 2.50 ~ 5.25; P < 0.00001; I2 = 70%), pregnancy rate (RR = 3.88; 95% CI: 1.94 ~ 7.79; P = 0.0001; I2 = 0%), count of births (SMD = 2.17; 95% CI: 1.31 ~ 3.04; P < 0.00001; I2 = 69%) and counts of different types of follicles. In addition, it elevates the level of AMH (SMD = 4.15; 95% CI: 2.75 ~ 5.54; P < 0.00001; I2 = 88%) and E2 (SMD = 2.88; 95% CI: 2.02 ~ 3.73; P < 0.00001; I2 = 80%) expression, while reducing FSH expression (SMD = -5.05; 95% CI: -6.60 ~ -3.50; P < 0.00001; I2 = 90%). Subgroup analysis indicates that the source of EVs, animal species, modeling method, administration route, and test timepoint affected efficacy. Trial sequential analysis showed that there was sufficient evidence to confirm the effects of stem cell-derived EVs on birth counts, ovarian weights, and follicle counts. However, the impact of stem cell-derived EVs on pregnancy rates needs to be further demonstrated through more animal experimental evidence. CONCLUSIONS Stem cell-derived EVs demonstrate safety and efficacy in treating POF animal models, with potential improvements in fertility outcomes. TRIAL REGISTRATION PROSPERO registration number: CRD42024509699.
Collapse
Affiliation(s)
- Yan Luo
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Medicine Eight-Year Program, Xiangya Hospital, Central South University, Changsha, China
| | - Jingjing Chen
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Jinyao Ning
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Medicine Eight-Year Program, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Sun
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Yitong Chai
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Medicine Eight-Year Program, Xiangya Hospital, Central South University, Changsha, China
| | - Fen Xiao
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Bixia Huang
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Ge Li
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Fen Tian
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Jie Hao
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Qiong Zhang
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Jing Zhao
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Yanping Li
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Hui Li
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China.
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China.
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
45
|
Wang YL, Yin LX, Li M. Assessment of left ventricular myocardial systolic dysfunction in premature ovarian insufficiency patients using echocardiographic layer-specific myocardial strain imaging. Echo Res Pract 2024; 11:20. [PMID: 39218983 PMCID: PMC11367989 DOI: 10.1186/s44156-024-00056-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Due to the lack of oestrogen, premature ovarian insufficiency (POI) is an independent risk factor for ischaemic heart disease and overall cardiovascular disease. This study aimed to apply layer-specific myocardial strain for early quantitative evaluation of subclinical left ventricular myocardial systolic function changes in patients with POI. METHODS Forty-eight newly diagnosed, untreated patients with POI (POI group) and fifty healthy female subjects matched for age, height and weight (control group) were enrolled. Standard transthoracic echocardiography was used to measure conventional parameters and layer-specific strain parameters.The layer-specific strain parameters included subendomyocardial global longitudinal strain (GLSendo), mid-layer myocardial global longitudinal strain (GLSmid), subepimyocardial global longitudinal strain (GLSepi), subendomyocardial global circumferential strain (GCSendo), mid-layer myocardial global circumferential strain (GCSmid), and subepimyocardial global circumferential strain (GCSepi). RESULTS There were no significant differences in age, body mass index (BMI), blood pressure, or left ventricular ejection fraction (LVEF) between the two groups. The end-diastolic interventricular septal thickness (IVST) was greater in the POI group (8.29 ± 1.32 vs. 7.66 ± 0.82, P = 0.008), and the POI group had lower E, E/A, and lateral e' (all P < 0.05). As for systolic functions,the POI group had lower GLSendo, GLSmid, GLSepi, GCSendo, GCSmid, and GCSepi (all P < 0.05).The intraobserver and interobserver coefficients of GLSendo, GLSmid, GLSepi, GCSendo, GCSmid, and GCSepi were greater than 0.900. CONCLUSIONS POI patients with normal LVEF may suffer from subclinical left ventricular myocardial systolic dysfunction. Echocardiography of layer-specific myocardial strain could more sensitively detect subclinical impairment of left ventricular systolic function in POI patients.
Collapse
Affiliation(s)
- Yu-Lin Wang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Li-Xue Yin
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Mei Li
- Department of Gynaecology and Obstetrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
46
|
Cao M, Yuan C, Chen X, He G, Chen T, Zong J, Shen C, Wang N, Zhao Y, Zhang B, Li C, Zhou X. METTL3 deficiency leads to ovarian insufficiency due to IL-1β overexpression in theca cells. Free Radic Biol Med 2024; 222:72-84. [PMID: 38825211 DOI: 10.1016/j.freeradbiomed.2024.05.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/20/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
Premature ovarian insufficiency (POI) is a clinical syndrome characterised by a decline in ovarian function in women before 40 years of age and is associated with oestradiol deficiency and a complex pathogenesis. However, the aetiology of POI is still unclear and effective preventative and treatment strategies are still lacking. Methyltransferase like 3 (METTL3) is an RNA methyltransferase that is involved in spermatogenesis, oocyte development and maturation, early embryonic development, and embryonic stem cell differentiation and formation, but its role in POI is unknown. In the present study, METTL3 deficiency in follicular theca cells was found to lead to reduced fertility in female mice, with a POI-like phenotype, and METTL3 knockout promoted ovarian inflammation. Further, a reduction in METTL3 in follicular theca cells led to a decrease in the m6A modification of pri-miR-21, which further reduced pri-miR-21 recognition and binding by DGCR8 proteins, leading to a decrease in the synthesis of mature miR-21-5p. Decrease of miR-21-5p promoted the secretion of interleukin-1β (IL-1β) from follicular theca cells. Acting in a paracrine manner, IL-1β inhibited the cAMP-PKA pathway and activated the NF-κB pathway in follicular granulosa cells. This activation increased the levels of reactive oxygen species in granulosa cells, causing disturbances in the intracellular Ca2+ balance and mitochondrial damage. These cellular events ultimately led to granulosa cell apoptosis and a decrease in oestradiol synthesis, resulting in POI development. Collectively, these findings reveal how METTL3 deficiency promotes the expression and secretion of IL-1β in theca cells, which regulates ovarian functions, and proposes a new theory for the development of POI disease.
Collapse
Affiliation(s)
- Maosheng Cao
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Chenfeng Yuan
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Xue Chen
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Guitian He
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Tong Chen
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Jinxin Zong
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Caomeihui Shen
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Nan Wang
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Yun Zhao
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Boqi Zhang
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Chunjin Li
- College of Animal Sciences, Jilin University, Changchun, 130062, China.
| | - Xu Zhou
- College of Animal Sciences, Jilin University, Changchun, 130062, China.
| |
Collapse
|
47
|
Tucker EJ, Sharp MF, Lokchine A, Bell KM, Palmer CS, Kline BL, Robevska G, van den Bergen J, Dulon J, Stojanovski D, Ayers KL, Touraine P, Crismani W, Jaillard S, Sinclair AH. Biallelic FANCA variants detected in sisters with isolated premature ovarian insufficiency. Clin Genet 2024; 106:321-335. [PMID: 38779778 DOI: 10.1111/cge.14543] [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: 12/07/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024]
Abstract
Premature ovarian insufficiency is a common form of female infertility affecting up to 4% of women and characterised by amenorrhea with elevated gonadotropin before the age of 40. Oocytes require controlled DNA breakage and repair for homologous recombination and the maintenance of oocyte integrity. Biallelic disruption of the DNA damage repair gene, Fanconi anemia complementation group A (FANCA), is a common cause of Fanconi anaemia, a syndrome characterised by bone marrow failure, cancer predisposition, physical anomalies and POI. There is ongoing dispute about the role of heterozygous FANCA variants in POI pathogenesis, with insufficient evidence supporting causation. Here, we have identified biallelic FANCA variants in French sisters presenting with POI, including a novel missense variant of uncertain significance and a likely pathogenic deletion that initially evaded detection. Functional studies indicated no discernible effect on DNA damage sensitivity in patient lymphoblasts. These novel FANCA variants add evidence that heterozygous loss of one allele is insufficient to cause DNA damage sensitivity and POI. We propose that intragenic deletions, that are relatively common in FANCA, may be missed without careful analysis, and could explain the presumed causation of heterozygous variants. Accurate variant curation is critical to optimise patient care and outcomes.
Collapse
Affiliation(s)
- Elena J Tucker
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael F Sharp
- DNA Repair and Recombination Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
- The Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Victoria, Australia
| | - Anna Lokchine
- CHU Rennes, INSERM, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S1085, Univ Rennes, Rennes, France
- Service de Cytogénétique et Biologie Cellulaire, CHU Rennes, Rennes, France
| | - Katrina M Bell
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Bioinformatics, Murdoch Children's Research Institute, Victoria, Australia
| | - Catherine S Palmer
- Department of Biochemistry and Pharmacology and The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Brianna L Kline
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Gorjana Robevska
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Jocelyn van den Bergen
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Jérôme Dulon
- Department of Endocrinology and Reproductive Medicine, AP-HP, Sorbonne University, Paris, France
| | - Diana Stojanovski
- Department of Biochemistry and Pharmacology and The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Katie L Ayers
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Philippe Touraine
- Department of Endocrinology and Reproductive Medicine, AP-HP, Sorbonne University, Paris, France
| | - Wayne Crismani
- DNA Repair and Recombination Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Sylvie Jaillard
- CHU Rennes, INSERM, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S1085, Univ Rennes, Rennes, France
- Service de Cytogénétique et Biologie Cellulaire, CHU Rennes, Rennes, France
| | - Andrew H Sinclair
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
48
|
Sun H, Qi Q, Pan X, Zhou J, Wang J, Li L, Li D, Wang L. Bu-Shen-Ning-Xin decoction inhibits macrophage activation to ameliorate premature ovarian insufficiency-related osteoimmune disorder via FSH/FSHR pathway. Drug Discov Ther 2024; 18:106-116. [PMID: 38631868 DOI: 10.5582/ddt.2024.01006] [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: 04/19/2024]
Abstract
Limited studies are associated with premature ovarian insufficiency (POI)-related osteoimmune disorder currently. Bu-Shen-Ning-Xin decoction (BSNXD) displayed a favorable role in treating postmenopausal osteoporosis. However, its impact on the POI-related osteoimmune disorder remains unclear. The study primarily utilized animal experiments and network pharmacology to investigate the effects and underlying mechanisms of BSNXD on the POI-related osteoimmune disorder. First, a 4-vinylcyclohexene dioxide (VCD)-induced POI murine model was conducted to explore the therapeutical action of BSNXD. Second, we analyzed the active compounds of BSNXD and predicted their potential mechanisms for POI-related osteoimmune disorder via network pharmacology, further confirmed by molecular biology experiments. The results demonstrated that VCD exposure led to elevated follicle-stimulating hormone (FSH) levels, a 50% reduction in the primordial follicles, bone microstructure changes, and macrophage activation, indicating an osteoimmune disorder. BSNXD inhibited macrophage activation and osteoclast differentiation but did not affect serum FSH and estradiol levels in the VCD-induced POI model. Network pharmacology predicted the potential mechanisms of BSNXD against the POI-related osteoimmune disorder involving tumor necrosis factor α and MAPK signaling pathways, highlighting BSNXD regulated inflammation, hormone, and osteoclast differentiation. Further experiments identified BSNXD treatment suppressed macrophage activation via downregulating FSH receptor (FSHR) expression and inhibiting the phosphorylation of ERK and CCAAT enhancer binding proteins β. In conclusion, BSNXD regulated POI-related osteoimmune disorder by suppressing the FSH/FSHR pathway to reduce macrophage activation and further inhibiting osteoclastogenesis.
Collapse
Affiliation(s)
- Hongmei Sun
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
- Hexi University, Zhangye, Gansu, China
| | - Qing Qi
- Wuhan Business University, Wuhan, Hubei, China
| | - Xinyao Pan
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Zhou
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Lisha Li
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Dajing Li
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Ling Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| |
Collapse
|
49
|
Bakhshalizadeh S, Afkhami F, Bell KM, Robevska G, van den Bergen J, Cronin S, Jaillard S, Ayers KL, Kumar P, Siebold C, Xiao Z, Tate EW, Danaei S, Farzadi L, Shahbazi S, Sinclair AH, Tucker EJ. Diverse genetic causes of amenorrhea in an ethnically homogeneous cohort and an evolving approach to diagnosis. Mol Cell Endocrinol 2024; 587:112212. [PMID: 38521400 DOI: 10.1016/j.mce.2024.112212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/12/2024] [Accepted: 03/16/2024] [Indexed: 03/25/2024]
Abstract
RESEARCH QUESTION Premature ovarian insufficiency (POI) is characterised by amenorrhea associated with elevated follicle stimulating hormone (FSH) under the age of 40 years and affects 1-3.7% women. Genetic factors explain 20-30% of POI cases, but most causes remain unknown despite genomic advancements. DESIGN We used whole exome sequencing (WES) in four Iranian families, validated variants via Sanger sequencing, and conducted the Acyl-cLIP assay to measure HHAT enzyme activity. RESULTS Despite ethnic homogeneity, WES revealed diverse genetic causes, including a novel homozygous nonsense variant in SYCP2L, impacting synaptonemal complex (SC) assembly, in the first family. Interestingly, the second family had two independent causes for amenorrhea - the mother had POI due to a novel homozygous loss-of-function variant in FANCM (required for chromosomal stability) and her daughter had primary amenorrhea due to a novel homozygous GNRHR (required for gonadotropic signalling) frameshift variant. WES analysis also provided cytogenetic insights. WES revealed one individual was in fact 46, XY and had a novel homozygous missense variant of uncertain significance in HHAT, potentially responsible for complete sex reversal although functional assays did not support impaired HHAT activity. In the remaining individual, WES indicated likely mosaic Turners with the majority of X chromosome variants having an allelic balance of ∼85% or ∼15%. Microarray validated the individual had 90% 45,XO. CONCLUSIONS This study demonstrates the diverse causes of amenorrhea in a small, isolated ethnic cohort highlighting how a genetic cause in one individual may not clarify familial cases. We propose that, in time, genomic sequencing may become a single universal test required for the diagnosis of infertility conditions such as POI.
Collapse
Affiliation(s)
- Shabnam Bakhshalizadeh
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Fateme Afkhami
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Katrina M Bell
- Department of Bioinformatics, Murdoch Children's Research Institute, Melbourne, Australia
| | | | | | - Sara Cronin
- Cyto-Molecular Diagnostic Research Laboratory, Victorian Clinical Genetics Services and Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052, Victoria, Australia
| | - Sylvie Jaillard
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France; CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, F-35033, Rennes, France
| | - Katie L Ayers
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Pramod Kumar
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Zhangping Xiao
- Department of Chemistry, Imperial College London, 82 Wood Lane, London, W12 0BZ, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, 82 Wood Lane, London, W12 0BZ, UK
| | - Shahla Danaei
- Department of Obstetrics and Gynecology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Laya Farzadi
- Department of Obstetrics and Gynecology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Shahbazi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Andrew H Sinclair
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Elena J Tucker
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
50
|
Jones AR, Enticott J, Ebeling PR, Mishra GD, Teede HT, Vincent AJ. Bone health in women with premature ovarian insufficiency/early menopause: a 23-year longitudinal analysis. Hum Reprod 2024; 39:1013-1022. [PMID: 38396142 PMCID: PMC11063537 DOI: 10.1093/humrep/deae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
STUDY QUESTION What is the frequency of, and predictors for, osteoporosis, fractures, and osteoporosis management (investigation, treatment) in women with premature ovarian insufficiency (POI; menopause <40 years) and early menopause (EM; menopause 40-44years)? SUMMARY ANSWER Over the 23-year follow-up duration, at a mean age of 68 years, women with POI/EM had higher osteoporosis/fracture risk and prevalence, higher osteoporosis screening and anti-osteoporosis medication use compared to women with usual age menopause; increasing age was predictive of increased risk of osteoporosis/fracture and menopause hormone therapy (MHT) prior to or at study entry (aged 45-50 years) was protective. WHAT IS KNOWN ALREADY Women with POI/EM have increased risk of osteoporosis and fractures with limited data regarding risk factors for reduced bone density and fractures. Clinical guidelines recommend screening with dual X-ray absorptiometry (DXA) and treatment with MHT for most women with POI/EM to reduce osteoporosis and fracture risk; however, studies indicate gaps in osteoporosis knowledge, guideline uptake, and management adherence by clinicians and women. STUDY DESIGN, SIZE, DURATION The Australian Longitudinal Study on Women's Health is a prospective longitudinal study of Australian women. This study uses the cohort of women born between 1946 and 1951, surveyed nine times between 1996 and 2019. Data from the Australian administrative health records, including hospital admissions data (fractures, osteoporosis), Medicare Benefits Schedule (DXA), and the Pharmaceutical Benefits Scheme (PBS; MHT, anti-osteoporosis medication, available only from 2002) were linked to survey data. PARTICIPANTS/MATERIALS, SETTING, METHODS Survey respondents with self-reported age of menopause were included. POI/EM was defined as menopause <45 years. T-test or chi-square were used for comparisons at baseline (P < 0.05 indicates significance). Generalized estimating equations for panel data explored predictors for the longitudinal outcomes of osteoporosis, fractures, DXA rates, MHT use, and anti-osteoporosis medication (in women with osteoporosis/fracture, from Survey 4 onwards only). Univariable regression was performed, and variables retained where P < 0.2, to form the multivariable model, and bootstrapping with 100 repetitions at 95% sampling of the original dataset to ensure robustness of results. MAIN RESULTS AND THE ROLE OF CHANCE Eight thousand six hundred and three women were included: 610 (7.1%) with POI/EM. Mean (SD) baseline age was 47.6 (1.45) years in the entire cohort and mean (SD) age of menopause was 38.2 (7.95) and 51.3 (3.04) years in women with POI/EM and usual age menopause, respectively (P < 0.001). Over the 23 years, of women with POI/EM, 303 (49.7%) had osteoporosis/fractures, 421 (69.0%) had DXA screening, 474 ever used MHT (77.7%), and 116 (39.1%) of those with osteoporosis/fractures used anti-osteoporosis medication. Of women with usual age menopause, 2929 (36.6%) had osteoporosis/fractures, 4920 (61.6%) had DXA screening, 4014 (50.2%) used MHT, and 964 (33.0%) of those with osteoporosis/fractures used anti-osteoporosis medication. Compared to women with menopause at age ≥45 years and after adjusting for other risk factors, women with POI/EM had increased risk of osteoporosis (odds ratio [OR] 1.37; 95% CI 1.07-1.77), fractures (OR 1.45; 1.15-1.81), DXA testing (OR 1.64; 1.42-1.90), MHT use (OR 6.87; 5.68-8.30), and anti-osteoporosis medication use (OR 1.50; 1.14-1.98). In women with POI/EM women, increasing age was associated with greater risk of osteoporosis/fracture (OR 1.09; 1.08-1.11), and MHT prior to or at study entry (aged 45-50 years), was protective (OR 0.65, 0.45-0.96). In women with POI/EM, age (OR 1.11; 1.10-1.12), fractures (OR 1.80, 1.38-2.34), current smoking (OR 0.60; 0.43-0.86), and inner (OR 0.68; 0.53-0.88) or outer regional (OR 0.63; 0.46-0.87) residential location were associated with DXA screening. In women with POI/EM, increasing age (OR 1.02; 1.01-1.02), and currently consuming alcohol (OR 1.17; 1.06-1.28), was associated with having ever used MHT. In the 299 women with POI/EM and osteoporosis/fractures, only 39.1% ever received treatment with an anti-osteoporosis medication. Increasing age (OR 1.07; 1.04-1.09) and lower BMI (OR 0.95; 0.92-0.98) were associated with greater likelihood of treatment with anti-osteoporosis medication. LIMITATIONS, REASONS FOR CAUTION Survey data including age of menopause were self-reported by participants; fracture questions were not included in the 2001 survey, and location or level of trauma of self-reported fractures was not asked. Additional risk/protective factors such as vitamin D status, calcium intake, and exercise were not able to be included. Due to sample size, POI and EM were combined for all analyses, and we were unable to differentiate between causes of POI/EM. PBS data were only available from 2004, and hospital admissions data were state-based, with all of Australia were only available from 2007. WIDER IMPLICATIONS OF THE FINDINGS This study supports previous literature indicating increased risk of osteoporosis and fractures in women with POI, and adds evidence for women with POI/EM, where there was a relative paucity of data. This is the first study to analyse a variety of clinical and demographic risk factors for osteoporosis and fractures in women with POI/EM, as well as analysing investigation and treatment rates. In these women, using MHT prior to or at study entry, aged 45-50 years, was protective for osteoporosis/fractures; however, having ever used MHT was not, highlighting the importance of early treatment with MHT in these women to preserve bone strength. Although women with POI/EM and osteoporosis or fractures were more likely to use anti-osteoporosis medications than those with usual age menopause, overall treatment rates are low at <40%, demonstrating a significant treatment gap that should be addressed to reduce future fracture risk. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by The Australian NHMRC Centre of Research Excellence Women's Health in Reproductive Life (CRE-WHIRL, project number APP1171592). A.R.J. is the recipient of a National Health and Medical Research Council post-graduate research scholarship (grant number 1169192). P.R.E. is supported by a National Health and Medical Research Council grant 1197958. P.R.E. reports grants paid to their institution from Amgen, Sanofi, and Alexion, honoraria from Amgen paid to their institution, and honoraria from Alexion and Kyowa-Kirin. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- A R Jones
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, VIC, Australia
- Department of Endocrinology, Monash Health, Melbourne, VIC, Australia
| | - J Enticott
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, VIC, Australia
| | - P R Ebeling
- Department of Endocrinology, Monash Health, Melbourne, VIC, Australia
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
| | - G D Mishra
- Australian Women and Girls’ Health Research Centre, School of Public Health, University of Queensland, Brisbane, QLD, Australia
| | - H T Teede
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, VIC, Australia
- Department of Endocrinology, Monash Health, Melbourne, VIC, Australia
| | - A J Vincent
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, VIC, Australia
- Department of Endocrinology, Monash Health, Melbourne, VIC, Australia
| |
Collapse
|