1
|
Karaviti E, Karaviti D, Kani ER, Chatziandreou E, Paschou SA, Psaltopoulou T, Kalantaridou S, Lambrinoudaki I. The role of anti-Müllerian hormone: insights into ovarian reserve, primary ovarian insufficiency, and menopause prediction. Endocrine 2025:10.1007/s12020-025-04265-0. [PMID: 40410629 DOI: 10.1007/s12020-025-04265-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 04/28/2025] [Indexed: 05/25/2025]
Abstract
This review highlights the role of Anti-Müllerian Hormone (AMH) in ovarian insufficiency and as a predictor of menopause. AMH, produced by granulosa cells in growing follicles, is a key marker of ovarian reserve, reflecting the remaining pool of viable follicles. In cases of primary ovarian insufficiency (POI), AMH levels are significantly reduced, aiding in diagnosis and distinguishing POI from other causes of amenorrhea. AMH levels below 8 pmol/L have shown high sensitivity (85%) and specificity (100%) for diagnosing POI in women with secondary oligomenorrhea. Regarding ovarian aging, AMH declines steadily with age, serving as a reliable predictor of menopausal timing. AMH levels are linked to menopausal symptoms, particularly vasomotor symptoms like hot flashes and their severity. However, its reliability for diagnosing menopause is inconsistent, especially in younger populations or when determining the exact onset. AMH levels can predict an earlier onset of menopause with limited sensitivity and specificity, particularly when using age-specific concentrations, as lower age-specific AMH levels are associated with an earlier menopause onset. Tracking AMH over time can improve the prediction of menopause. The accuracy of AMH measurements can be enhanced when considered alongside other hormonal markers or clinical symptoms. In polycystic ovary syndrome (PCOS), elevated AMH levels suggest a delayed onset of menopause, indicating an approximately two-year longer reproductive lifespan compared to women without PCOS (mean menopause age: 51.4 years in PCOS cases vs. 49.7 years in healthy controls). In endometriosis, AMH levels generally decline after surgery; however, they remain stable after chemotherapy, even years later, indicating that the decline in ovarian reserve may not be significantly affected.
Collapse
Affiliation(s)
- Eleftheria Karaviti
- Endocrine Unit and Diabetes Centre, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitra Karaviti
- Endocrine Unit and Diabetes Centre, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni-Rafaela Kani
- Endocrine Unit and Diabetes Centre, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathia Chatziandreou
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Stavroula A Paschou
- Endocrine Unit and Diabetes Centre, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Theodora Psaltopoulou
- Endocrine Unit and Diabetes Centre, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sophia Kalantaridou
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Irene Lambrinoudaki
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
2
|
Parent AS, Damdimopoulou P, Johansson HKL, Bouftas N, Draskau MK, Franssen D, Fudvoye J, van Duursen M, Svingen T. Endocrine-disrupting chemicals and female reproductive health: a growing concern. Nat Rev Endocrinol 2025:10.1038/s41574-025-01131-x. [PMID: 40404936 DOI: 10.1038/s41574-025-01131-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/06/2025] [Indexed: 05/24/2025]
Abstract
Female fertility and reproductive health depend on a series of developmental steps from embryogenesis through puberty, in addition to the proper functioning of the reproductive system in adulthood. Two important steps are the establishment of the ovarian reserve and development of the hypothalamic-pituitary-ovarian axis. During reproductive years, maintaining an adequate ovarian reserve of follicles as well as balanced neuroendocrine control of reproductive organs is crucial for fertility. Dysregulation of either of these events, during development or in adulthood, can lead to reproductive disorders. Over the past five decades, human fertility rates have declined, whereas the incidence of female reproductive disorders has risen, trends partially linked to environmental factors such as exposure to endocrine-disrupting chemicals (EDCs). Here we outline epidemiological and mechanistic evidence for how EDCs affect the ovarian reserve during early development, its maintenance during adulthood and the establishment of the hypothalamic-pituitary control of puberty and ovulation. Our Review not only reveals strong support for the role of EDC exposure in the development of female reproductive disorders such as abnormal puberty, impaired fertility, premature menopause or polycystic ovarian syndrome, but also highlights knowledge gaps, including the difficulty to prove causality between exposure and human disease manifestation.
Collapse
Affiliation(s)
- Anne-Simone Parent
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liege, Liege, Belgium.
- Department of Paediatrics, University Hospital Liege, Liege, Belgium.
| | - Pauliina Damdimopoulou
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | | | - Nora Bouftas
- Environmental Health and Toxicology, Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Monica K Draskau
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Delphine Franssen
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liege, Liege, Belgium
| | - Julie Fudvoye
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liege, Liege, Belgium
- Department of Paediatrics, University Hospital Liege, Liege, Belgium
| | - Majorie van Duursen
- Environmental Health and Toxicology, Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
3
|
Mavaddat N, Frost D, Zhao E, Barnes DR, Ahmed M, Barwell J, Brady AF, Brennan P, Conti H, Cook J, Copeland H, Davidson R, Donaldson A, Douglas E, Gallagher D, Hart R, Izatt L, Kemp Z, Lalloo F, Miedzybrodzka Z, Morrison PJ, Murray JE, Murray A, Musgrave H, Searle C, Side L, Snape K, Tripathi V, Walker L, Archer S, Evans DG, Tischkowitz M, Antoniou AC, Easton DF. Distribution of age at natural menopause, age at menarche, menstrual cycle length, height and BMI in BRCA1 and BRCA2 pathogenic variant carriers and non-carriers: results from EMBRACE. Breast Cancer Res 2025; 27:87. [PMID: 40399999 PMCID: PMC12093752 DOI: 10.1186/s13058-025-02030-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/20/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Carriers of germline pathogenic variants (PVs) in the BRCA1 and BRCA2 genes are at higher risk of developing breast and ovarian cancer than the general population. It is unclear if these PVs influence other breast or ovarian cancer risk factors, including age at menopause (ANM), age at menarche (AAM), menstrual cycle length, BMI or height. There is a biological rationale for associations between BRCA1 and BRCA2 PVs and reproductive traits, for example involving DNA damage and repair mechanisms. The evidence for or against such associations is limited. METHODS We used data on 3,046 BRCA1 and 3,264 BRCA2 PV carriers, and 2,857 non-carrier female relatives of PV carriers from the Epidemiological Study of Familial Breast Cancer (EMBRACE). Associations between ANM and PV carrier status was evaluated using linear regression models allowing for censoring. AAM, menstrual cycle length, BMI, and height in carriers and non-carriers were compared using linear and multinomial logistic regression. Analyses were adjusted for potential confounders, and weighted analyses carried out to account for non-random sampling with respect to cancer status. RESULTS No statistically significant difference in ANM between carriers and non-carriers was observed in analyses accounting for censoring. Linear regression effect sizes for ANM were -0.002 (95%CI: -0.401, 0.397) and -0.172 (95%CI: -0.531, 0.188), for BRCA1 and BRCA2 PV carriers respectively, compared with non-carrier women. The distributions of AAM, menstrual cycle length and BMI were similar between PV carriers and non-carriers, but BRCA1 PV carriers were slightly taller on average than non-carriers (0.5 cm difference, p = 0.003). CONCLUSION Information on the distribution of cancer risk factors in PV carriers is needed for incorporating these factors into multifactorial cancer risk prediction algorithms. Contrary to previous reports, we found no evidence that BRCA1 or BRCA2 PV are associated with hormonal or anthropometric factors, except for a weak association with height. We highlight methodological considerations and data limitations inherent in studies aiming to address this question.
Collapse
Affiliation(s)
- Nasim Mavaddat
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK.
| | - Debra Frost
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Emily Zhao
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Daniel R Barnes
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Munaza Ahmed
- North East Thames Regional Clinical Genetics Service, Great Ormond Street Hospital, London, UK
| | - Julian Barwell
- Leicestershire, Northamptonshire and Rutland Clinical Genetics Service, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Angela F Brady
- North West Thames Regional Genetics Service, London North West University Healthcare NHS Trust, London, UK
| | - Paul Brennan
- Northern Genetics Service, Newcastle Upon Tyne NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Hector Conti
- All Wales Medical Genomics Services, Wrexham Maelor Hospital, Wrexham, UK
| | - Jackie Cook
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital, Sheffield, UK
| | - Harriet Copeland
- Department Clinical Genetics, Royal Devon University Healthcare NHS Foundation Trust, Exeter, Devon, UK
| | - Rosemarie Davidson
- Department of Clinical Genetics, South Glasgow University Hospitals, Glasgow, UK
| | - Alan Donaldson
- Clinical Genetics Department, St Michael's Hospital, Bristol, UK
| | - Emma Douglas
- West Midlands Regional Clinical Genetics Service, Birmingham Women's Hospital, Birmingham, UK
| | - David Gallagher
- Trinity St Jame's Cancer Institute, Cancer Genetics Service, Dublin, Ireland
| | - Rachel Hart
- Liverpool Women's Hospital Cheshire and Merseyside Genetics, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | - Louise Izatt
- Clinical Genetics, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Zoe Kemp
- Royal Marsden Hospital, NHS Trust, London, England, UK
| | - Fiona Lalloo
- Clinical Genetics Service, Manchester Centre for Genomic Medicine, Manchester University Hospitals Foundation Trust, Manchester, UK
| | - Zosia Miedzybrodzka
- NHS Grampian, North of Scotland Regional Genetics Service, Aberdeen, Scotland, UK
| | - Patrick J Morrison
- Belfast Health and Social Care Trust, Clinical Genetics Service, Belfast, Northern Ireland, UK
| | - Jennie E Murray
- South East Scotland Clinical Genetics Service, Western General Hospital, Edinburgh, UK
| | - Alex Murray
- All Wales Medical Genomics Service, Wales Genomic Health Centre, Cardiff, UK
| | - Hannah Musgrave
- Leeds Genomic Medicine Service, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Claire Searle
- Department of Clinical Genetics, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Lucy Side
- University Hospital Southampton NHS Trust and Princess Anne Hospital, Southampton, UK
| | - Katie Snape
- Medical Genetics Unit, St George's, University of London, London, UK
| | - Vishakha Tripathi
- Clinical Genetics, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Lisa Walker
- Oxford Centre for Genomic Medicine, Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Stephanie Archer
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - D Gareth Evans
- Genomic Medicine, Manchester Academic Health Sciences Centre, Division of Evolution, Infection and Genomic Science, University of Manchester, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Marc Tischkowitz
- Department of Genomic Medicine, Cambridge Biomedical Research Centre, National Institute for Health Research, University of Cambridge, Cambridge, UK
| | - Antonis C Antoniou
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| |
Collapse
|
4
|
Kwon SA, Cho YS. Identification of loci associated with women's reproductive traits and exploration of a shared genetic basis with obesity. Hum Genomics 2025; 19:58. [PMID: 40394640 PMCID: PMC12093848 DOI: 10.1186/s40246-025-00773-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND The timing of menarche and menopause significantly affects women's health, with influences on cancer, cardiovascular disease, obesity, type 2 diabetes, and psychosocial problems. In addition, observational studies have reported that ages at menarche (AAM) and natural menopause (ANM) are correlated with obesity. To understand the genetic bases of these reproductive traits, we conducted a genome-wide association study (GWAS) of AAM and ANM in the Korean population. We also investigated the genetic correlation and causal relationship to explore the shared genetic architecture between reproductive traits and obesity in women. RESULTS Our GWA analyses of 45,608 and 21,599 adult women identified two and six genome-wide significant associations (P-value < 5 × 10- 8) for AAM and ANM, respectively. Although most of the loci that we detected have been reported in previous studies, we have newly linked the JHY locus containing the SNP rs11605693 to AAM. Leveraging the GWAS results, we tested the shared genetic basis underlying AAM and ANM, which appear to be closely related to female hormone activity. Linkage disequilibrium score regression (LDSC) analysis did not identify a significant genetic correlation between the two traits. Our LDSC analyses indicated that AAM was inversely correlated with two obesity traits, body mass index (BMI) and waist circumference (WC). However, Mendelian randomization (MR) analyses did not provide evidence of a causal relationship between AAM and obesity traits. CONCLUSIONS Overall, our study provides insights into the genetic architecture of women's reproductive traits and the shared genetic basis between AAM and obesity. Our MR analyses suggest that the genetic correlation between AAM and obesity traits results from the direct effects of genetic variants on both traits rather than a causal relationship between them.
Collapse
Affiliation(s)
- Seong-Ah Kwon
- Department of Biomedical Science, Hallym University, Chuncheon, 24252, Gangwon State, Republic of Korea
| | - Yoon Shin Cho
- Department of Biomedical Science, Hallym University, Chuncheon, 24252, Gangwon State, Republic of Korea.
- Department of Neuroscience, Hallym University College of Medicine, Chuncheon, 24252, Gangwon State, Republic of Korea.
- GenoMax Co., Ltd., Life Science Building 8406, Hallymdaehakgil 1, Chuncheon, Gangwon State, Republic of Korea.
| |
Collapse
|
5
|
Wang X, Mao R, Long R, Gao L, Wang M, Zhou J, Qian K, Zhu L, Jin L. Identification of potential therapeutic targeting in ovarian aging from genetic screening with clinical validation. J Assist Reprod Genet 2025:10.1007/s10815-025-03490-w. [PMID: 40343599 DOI: 10.1007/s10815-025-03490-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/16/2025] [Indexed: 05/11/2025] Open
Abstract
PURPOSE To screen drug targets of ovarian aging from a genetic perspective. METHODS Systematic analyses were conducted with cis-expression quantitative trait loci data of druggable genes extracted as instrument variables. Summary statistics were from large genome-wide association studies for age at menopause. The following colocalization analysis was utilized to examine whether identified genes and ovarian aging shared causal variants. Furthermore, clinical validation was conducted by comparing expression of identified genes in granulosa cells from women with normal or diminished ovarian reserve (DOR) who went through in vitro fertilization (IVF) and by evaluating correlation of targeted gene expression with ovarian function and IVF outcomes. Moreover, single-nuclear RNA (snRNA) seq and drug database were analyzed to find target cells within the ovary and potential drugs targeting identified genes. RESULTS Systematic analyses identified five therapeutic targets of ovarian aging, including four protective factors (BRCA1, KLHL18, PNP, SRPK1) and one risk factor (PDIA3). The change in expression level of four protective factors has been verified in clinical validation. Particularly, both BRCA1 and SRPK1 have been downregulated among advanced-aged women with DOR and were positively correlated with anti-Müllerian hormone and antral follicle count. Specific target cells and potential small molecule targeted drugs of these genes were identified through snRNA analysis and searching in the drug database. CONCLUSIONS By systematic genetic analyses combined with clinical validation, we identified five potential druggable genes for ovarian aging, providing theoretical basis and promising direction of therapeutic genetic targets for ovarian aging in the future.
Collapse
Affiliation(s)
- Xiangfei Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Ruolin Mao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Rui Long
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Limin Gao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Meng Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Juepu Zhou
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Kun Qian
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| | - Lixia Zhu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| | - Lei Jin
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| |
Collapse
|
6
|
Chen C, Gao D, Yue H, Wang H, Qu R, Hu X, Luo L. Predicting breast cancer prognosis based on a novel pathomics model through CHEK1 expression analysis using machine learning algorithms. PLoS One 2025; 20:e0321717. [PMID: 40344565 PMCID: PMC12064205 DOI: 10.1371/journal.pone.0321717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 03/05/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Checkpoint kinase 1 (CHEK1) is often overexpressed in solid tumors. Nonetheless, the prognostic significance of CHEK1 in breast cancer (BrC) remains unclear. This study used pathomics leverages machine learning to predict BrC prognosis based on CHEK1 gene expression.. METHODS Initially, hematoxylin-eosin (H&E)-stained images obtained from The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) were segmented using Otsu's method. Further, the sub-image features were extracted using machine learning algorithms based on PyRadiomics, mRMRe, and Gradient Boosting Machine (GBM). The predicted CHEK1 expression levels were represented as the pathomics score (PS) and validated using the corresponding RNA-seq data. The prognostic significance of both CHEK1 and PS was evaluated using Kaplan-Meier (KM), and univariate and multivariate Cox regression. The model was assessed by comparing CHEK1 expression by immunohistochemistry (IHC) with PS in BrC tissue microarray (TMA). RESULTS A 633 × 10 sub-image set was eligible for training and a 158 × 10 set for validation. 1,488 features were extracted and 8 recursive feature elimination (RFE)-screened features were used to generate the model. A high PS was associated with CHEK1 overexpression, significantly correlating with survival outcomes, especially within 96 months post-diagnosis. Further, patients with high PS responded to anti-programmed cell death protein 1 (anti-PD-1) and anti-cytotoxic T lymphocyte antigen-4 (anti-CTLA4) treatments. In TMA validation, the IHC analysis estimated that high PS similarly predicted poorer prognosis and correlated with higher CHEK1 expression. CONCLUSIONS The novel pathomics model reliably predicted CHEK1 expression using machine learning algorithms, which might provide potential clinical utility for prognosis and treatment guidance.
Collapse
Affiliation(s)
- Chen Chen
- Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Dan Gao
- Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Huan Yue
- Clinical Laboratory, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Huijing Wang
- Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Rui Qu
- Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Xiaochi Hu
- Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Libo Luo
- Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| |
Collapse
|
7
|
Hudobenko J, Di Gesù CM, Mooz PR, Petrosino J, Putluri N, Ganesh BP, Rebeles K, Blixt FW, Venna VR, McCullough LD. Maternal dysbiosis produces long-lasting behavioral changes in offspring. Mol Psychiatry 2025; 30:1847-1858. [PMID: 39443733 PMCID: PMC12014497 DOI: 10.1038/s41380-024-02794-0] [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: 02/27/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
Advanced maternal age (AMA) is defined as a pregnancy in a woman older than 35 years of age. AMA increases the risk for both maternal and neonatal complications, including miscarriage and stillbirth. AMA has also been linked to neurodevelopmental and neuropsychiatric disorders in the offspring. Recent studies have found that age-associated compositional shifts in the gut microbiota contribute to altered microbial metabolism and enhanced inflammation in the host. We investigated the specific contribution of the maternal microbiome on pregnancy outcomes and offspring behavior by recolonizing young female mice with aged female microbiome prior to pregnancy. We discovered that pre-pregnancy colonization of young dams with microbiome from aged female donors significantly increased fetal loss. There were significant differences in the composition of the gut microbiome in pups born from dams recolonized with aged female biome that persisted through middle age. Offspring born from dams colonized with aged microbiome also had significant changes in levels of neurotransmitters and metabolites in the blood and the brain. Adult offspring from dams colonized with an aged microbiome displayed persistent depressive- and anxiety-like phenotypes. Collectively, these results demonstrate that age-related changes in the composition of the maternal gut microbiome contribute to chronic alterations in the behavior and physiology of offspring. This work highlights the potential of microbiome-targeted approaches, even prior to birth, may reduce the risk of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jacob Hudobenko
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Claudia M Di Gesù
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Patrick R Mooz
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
- UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Nagireddy Putluri
- Dan L. Duncan Comprehensive Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
- UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristen Rebeles
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
| | - Frank W Blixt
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
| | - Venugopal R Venna
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA.
- UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
8
|
Wu C, Chen D, Stout MB, Wu M, Wang S. Hallmarks of ovarian aging. Trends Endocrinol Metab 2025; 36:418-439. [PMID: 40000274 DOI: 10.1016/j.tem.2025.01.005] [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: 09/26/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025]
Abstract
Ovarian aging is considered to be the pacemaker of female aging, and is linked to various comorbidities such as osteoporosis, cardiovascular diseases, and cognitive decline. Many efforts have been made to determine the mechanisms underlying ovarian aging, but their potential to act as hallmarks to predict and intervene in this process currently remains unclear. In this review we propose nine hallmarks as common features of ovarian aging: genomic instability, telomere attrition, epigenetic alterations, impaired autophagy, cellular senescence, deregulated nutrient-sensing, mitochondrial dysfunction, oxidative stress, and chronic inflammation. Understanding the interaction between these hallmarks poses a significant challenge but may also pave the way to the identification of pharmaceutical targets that can attenuate ovarian aging.
Collapse
Affiliation(s)
- Chuqing Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China.
| |
Collapse
|
9
|
Zhao Z, Liang Y. Association between triglyceride-glucose index and phenotypic age acceleration: a cross-sectional study based on NHANES database. Front Physiol 2025; 16:1548690. [PMID: 40376115 PMCID: PMC12078276 DOI: 10.3389/fphys.2025.1548690] [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: 12/20/2024] [Accepted: 04/14/2025] [Indexed: 05/18/2025] Open
Abstract
Objective To investigate the association between triglyceride-glucose (TyG) index and phenotypic age acceleration (PhenoAgeAccel), given the emerging importance of biological aging as a health determinant and the role of insulin resistance in aging-related processes. Methods This cross-sectional study analyzed data from 13,291 adults aged ≥20 years in the National Health and Nutrition Examination Survey (1999-2010). The TyG index served as the exposure variable, calculated from fasting triglycerides and glucose levels. PhenoAgeAccel, derived from clinical biomarkers, was the outcome variable. Analyses adjusted for demographic, socioeconomic, and health-related covariates. Results A significant non-linear relationship was observed between TyG index and PhenoAgeAccel, with an inflection point at 9.60. In the fully adjusted model, each unit increase in TyG index was associated with 2.21 years increase in PhenoAgeAccel (95% CI: 1.99, 2.43). The association was stronger above the inflection point (β = 8.21, 95% CI: 7.59, 8.82) compared to below it (β = 0.56, 95% CI: 0.29, 0.83). Conclusion Higher TyG index levels are significantly associated with accelerated biological aging, particularly above a threshold of 9.60. These findings suggest the importance of metabolic health in biological aging processes and potential interventional strategies.
Collapse
Affiliation(s)
- Zhili Zhao
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Nursing, Sichuan University, Chengdu, China
| | - Yan Liang
- West China School of Nursing, Sichuan University, Chengdu, China
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Yi Y, Ma F, Jiao Y, Zhang R, Yi Y, Ma Y. Comparative efficacy of acupuncture therapies in premature ovarian failure: A systematic review and network meta-analysis. Complement Ther Med 2025; 89:103141. [PMID: 39922292 DOI: 10.1016/j.ctim.2025.103141] [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: 11/29/2024] [Revised: 01/12/2025] [Accepted: 01/31/2025] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVES Premature ovarian failure (POF) is becoming more common and has a major negative impact on women's mental and physical well-being. The use of acupuncture therapies to treat POF has gained popularity. However, the optimal treatment remains uncertain. This study aimed to systematically review the literature and conduct a network meta-analysis (NMA) to compare the efficacy of different acupuncture and related therapies and identify methodologic weaknesses in previous studies. DESIGN We systematically searched six databases from their inception to April 2024 for randomized controlled trials (RCTs) of acupuncture therapies for POF. We assessed methodological quality and risk of bias using the RoB 2.0 tool. The NMA was conducted using R and STATA software based on frequency theory, focusing on overall effectiveness rates and hormone levels. Additionally, we critically reviewed methodological limitations and potential biases in the studies included. RESULTS Thirty-seven eligible studies involving 2419 patients were included in this NMA. The NMA results indicated that moxibustion_catgut implantation at acupoint (MB_CIA) (P-score = 0.88; pooled mean difference (95 % CI): 22.07 (1.61-302.48)) showed the highest improvement in overall effectiveness, while warming acupuncture (WA) (0.95; -27.56 (-45.35 to -9.78)) improved follicle-stimulating hormone (FSH) levels best. WA_MB (0.85; -9.35 (-15.08 to -3.62)) and WA (P-score = 0.77) were most effective for luteinizing hormone (LH) and estradiol (E2) levels, respectively. Confidence in evidence ranged from moderate to very low, with low overall confidence. Key methodologic weaknesses included insufficient allocation concealment, lack of blinding, and small sample sizes. CONCLUSIONS Acupuncture therapies may effectively improve POF. MB_CIA, WA, and WA_MB seem to be the most effective. However, severe methodological constraints, such as insufficient randomization and a lack of blinding, may reduce trust in these results. To offer robust evidence, high-quality RCTs must overcome these limitations. SYSTEMATIC REVIEW REGISTRATION [PROSPERO], identifier [CRD42024550180].
Collapse
Affiliation(s)
- Yonghang Yi
- Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Fengjun Ma
- Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Yang Jiao
- Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Rongqi Zhang
- Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Yunhao Yi
- Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Yuxia Ma
- Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
11
|
Zhang F, Zhu M, Chen Y, Wang G, Yang H, Lu X, Li Y, Chang HM, Wu Y, Ma Y, Yuan S, Zhu W, Dong X, Zhao Y, Yu Y, Wang J, Mu L. Harnessing omics data for drug discovery and development in ovarian aging. Hum Reprod Update 2025; 31:240-268. [PMID: 39977580 DOI: 10.1093/humupd/dmaf002] [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: 09/29/2024] [Revised: 12/02/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Ovarian aging occurs earlier than the aging of many other organs and has a lasting impact on women's overall health and well-being. However, effective interventions to slow ovarian aging remain limited, primarily due to an incomplete understanding of the underlying molecular mechanisms and drug targets. Recent advances in omics data resources, combined with innovative computational tools, are offering deeper insight into the molecular complexities of ovarian aging, paving the way for new opportunities in drug discovery and development. OBJECTIVE AND RATIONALE This review aims to synthesize the expanding multi-omics data, spanning genome, transcriptome, proteome, metabolome, and microbiome, related to ovarian aging, from both tissue-level and single-cell perspectives. We will specially explore how the analysis of these emerging omics datasets can be leveraged to identify novel drug targets and guide therapeutic strategies for slowing and reversing ovarian aging. SEARCH METHODS We conducted a comprehensive literature search in the PubMed database using a range of relevant keywords: ovarian aging, age at natural menopause, premature ovarian insufficiency (POI), diminished ovarian reserve (DOR), genomics, transcriptomics, epigenomics, DNA methylation, RNA modification, histone modification, proteomics, metabolomics, lipidomics, microbiome, single-cell, genome-wide association studies (GWAS), whole-exome sequencing, phenome-wide association studies (PheWAS), Mendelian randomization (MR), epigenetic target, drug target, machine learning, artificial intelligence (AI), deep learning, and multi-omics. The search was restricted to English-language articles published up to September 2024. OUTCOMES Multi-omics studies have uncovered key mechanisms driving ovarian aging, including DNA damage and repair deficiencies, inflammatory and immune responses, mitochondrial dysfunction, and cell death. By integrating multi-omics data, researchers can identify critical regulatory factors and mechanisms across various biological levels, leading to the discovery of potential drug targets. Notable examples include genetic targets such as BRCA2 and TERT, epigenetic targets like Tet and FTO, metabolic targets such as sirtuins and CD38+, protein targets like BIN2 and PDGF-BB, and transcription factors such as FOXP1. WIDER IMPLICATIONS The advent of cutting-edge omics technologies, especially single-cell technologies and spatial transcriptomics, has provided valuable insights for guiding treatment decisions and has become a powerful tool in drug discovery aimed at mitigating or reversing ovarian aging. As technology advances, the integration of single-cell multi-omics data with AI models holds the potential to more accurately predict candidate drug targets. This convergence offers promising new avenues for personalized medicine and precision therapies, paving the way for tailored interventions in ovarian aging. REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Fengyu Zhang
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ming Zhu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yi Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Guiquan Wang
- Xiamen Key Laboratory of Reproduction and Genetics, Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinmei Lu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Li
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Yang Wu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yunlong Ma
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuai Yuan
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Wencheng Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jia Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liangshan Mu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Using genetics as a tool to understand female reproductive health better. Nat Med 2025; 31:1420-1421. [PMID: 40175740 DOI: 10.1038/s41591-025-03663-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
|
13
|
Zhang ZL, Ren ST, Yang WJ, Xu XW, Zhao SM, Fang KF, Lin Y, Yuan YY, Zhang XJ, Chen YQ, Xu W. AARS2-catalyzed lactylation induces follicle development and premature ovarian insufficiency. Cell Death Discov 2025; 11:209. [PMID: 40301335 PMCID: PMC12041370 DOI: 10.1038/s41420-025-02501-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 04/13/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
Lactate, a metabolite which is elevated in various developmental and pathological processes, exerts its signal through alanyl tRNA synthetases (AARS)-catalyzed protein lactylation. Herein, we report that elevated lactate and gain-of-function mitochondrial AARS (AARS2) mutations-induced hyper-lactylation promotes premature ovarian insufficiency (POI). Serum lactate is elevated in POI patients. POI-driving AARS2 mutations gain lactyltransferase activity. AARS2 lactylates and inactivates carnitine palmitoyl transferase 2 (CPT2), resulting in FFA accumulation that activates peroxisome proliferator-activated receptor γ (PPARγ), and potentiates follicle-stimulating hormone (FSH) to initiate follicle development. These, in synergy with the anabolites accumulation effects of AARS2, promoted lactylation-induced PDHA1 inactivation promote granular cell (GC) proliferation and primordial follicle development. GC-specific AARS2 overexpression does not affect primordial follicle number but speed up follicle depletion. AARS2 ablation or lactylation-inhibiting β-alanine treatments can prevent folliculogenesis and POI traits in mouse. These findings reveal that lactate signal drives follicle development, and inhibiting lactate signal could treat/prevent POI.
Collapse
Affiliation(s)
- Zhi-Ling Zhang
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic and Development of Complex Phenotypes, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shu-Ting Ren
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic and Development of Complex Phenotypes, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Wan-Jie Yang
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic and Development of Complex Phenotypes, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiao-Wen Xu
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic and Development of Complex Phenotypes, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shi-Min Zhao
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic and Development of Complex Phenotypes, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ke-Fei Fang
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic and Development of Complex Phenotypes, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yan Lin
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic and Development of Complex Phenotypes, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yi-Yuan Yuan
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic and Development of Complex Phenotypes, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiao-Jin Zhang
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic and Development of Complex Phenotypes, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yun-Qin Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Wei Xu
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic and Development of Complex Phenotypes, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- Shanghai Fifth People's Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Wang N, Yu H, Yin J, Yu X. pH-responsive nano-vaccine combined with anti-PD-1 antibodies for enhanced immunotherapy of breast cancer. Theranostics 2025; 15:6022-6043. [PMID: 40365283 PMCID: PMC12068292 DOI: 10.7150/thno.107200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Objective: This study aimed to investigate the therapeutic potential and underlying mechanisms of a novel pH-responsive nano-vaccine in combination with anti-Programmed Cell Death Protein 1 (PD-1) antibodies for the treatment of breast cancer (BC), with a focus on tumor growth inhibition, metastasis prevention, and immune microenvironment modulation. Methods: A pH-responsive amphiphilic diblock copolymer was synthesized using reversible addition-fragmentation chain transfer (RAFT) polymerization and conjugated with STING agonist ADU-S100 and mannose to specifically target dendritic cells (DCs). The nano-vaccine was further formulated with antigen peptides and polyethyleneimine (PEI) to enhance antigen delivery. Its particle size, stability, and surface charge were characterized using dynamic light scattering (DLS) and zeta potential analysis. In vitro, the immunostimulatory capacity of the nano-vaccine was evaluated via flow cytometry (FCM) analysis of DC activation markers. In vivo, mouse immune and tumor recurrence models were used to assess the its effects on T-cell activation, tumor suppression, and immune memory induction. The therapeutic efficacy of nano-vaccine/anti-PD-1 combination therapy was further assessed. Results: The nano-vaccine efficiently activated DCs and promoted antigen presentation, as indicated by increased CD80, CD86, and MHC-II expression in vitro. In mouse models, it effectively inhibited tumor growth, induced antigen-specific T-cell responses, and suppressed recurrent and metastatic tumor progression. The combination with anti-PD-1 antibodies further enhanced tumor control, immune cell infiltration, and survival rates compared to monotherapy. Conclusion: The pH-responsive nano-vaccine combined with anti-PD-1 antibodies showed remarkable synergistic effects in BC treatment, highlighting its potential to enhance immune checkpoint blockade therapy and offer a promising strategy for clinical applications in solid tumors.
Collapse
Affiliation(s)
- Ning Wang
- Department of Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Hong Yu
- Department of Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jianqiao Yin
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiaopeng Yu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
15
|
Mörseburg A, Zhao Y, Kentistou KA, Perry JRB, Ong KK, Day FR. Genetic determinants of proteomic aging. NPJ AGING 2025; 11:30. [PMID: 40287427 PMCID: PMC12033249 DOI: 10.1038/s41514-025-00205-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/21/2025] [Indexed: 04/29/2025]
Abstract
Changes in the proteome and its dysregulation have long been known to be a hallmark of aging. We derived a proteomic aging trait using data on 1459 plasma proteins from 44,435 UK Biobank individuals measured using an antibody-based assay. This metric is strongly associated with four age-related disease outcomes, even after adjusting for chronological age. Survival analysis showed that one-year older proteomic age, relative to chronological age, increases all-cause mortality hazard by 13 percent. We performed a genome-wide association analysis of proteomic age acceleration (proteomic aging trait minus chronological age) to identify its biological determinants. Proteomic age acceleration showed modest genetic correlations with four epigenetic clocks (Rg = 0.17 to 0.19) and telomere length (Rg = -0.2). Once we removed associations that were explained by a single pQTL, we were left with three signals mapping to BRCA1, POLR2A and TET2 with apparent widespread effects on plasma proteomic aging. Genetic variation at these three loci has been shown to affect other omics-related aging measures. Mendelian randomisation analyses showed causal effects of higher BMI and type 2 diabetes on faster proteomic age acceleration. This supports the idea that obesity and other features of metabolic syndrome have an adverse effect on the processes of human aging.
Collapse
Affiliation(s)
- Alexander Mörseburg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | - Yajie Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Katherine A Kentistou
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John R B Perry
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Felix R Day
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| |
Collapse
|
16
|
Allen-Brady K, Moore B, Verrilli LE, Alvord MA, Kern M, Camp N, Kelley K, Letourneau J, Cannon-Albright L, Yandell M, Johnstone EB, Welt CK. Breast Cancer Is Increased in Women With Primary Ovarian Insufficiency. J Clin Endocrinol Metab 2025; 110:e1678-e1686. [PMID: 38996041 PMCID: PMC12012772 DOI: 10.1210/clinem/dgae480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
CONTEXT DNA damage/repair gene variants are associated with both primary ovarian insufficiency (POI) and cancer risk. OBJECTIVE We hypothesized that a subset of women with POI and family members would have increased risk for cancer. DESIGN Case-control population-based study using records from 1995 to 2022. SETTING Two major Utah academic health care systems serving 85% of the state. SUBJECTS Women with POI (n = 613) were identified using International Classification of Diseases codes and reviewed for accuracy. Relatives were linked using the Utah Population Database. INTERVENTION Cancer diagnoses were identified using the Utah Cancer Registry. MAIN OUTCOME MEASURES The relative risk of cancer in women with POI and relatives was estimated by comparison to population rates. Whole genome sequencing was performed on a subset of women. RESULTS Breast cancer was increased in women with POI (OR, 2.20; 95% CI, 1.30-3.47; P = .0023) and there was a nominally significant increase in ovarian cancer. Probands with POI were 36.5 ± 4.3 years and 59.5 ± 12.7 years when diagnosed with POI and cancer, respectively. Causal and candidate gene variants for cancer and POI were identified. Among second-degree relatives of these women, there was an increased risk of breast (OR, 1.28; 95% CI, 1.08-1.52; P = .0078) and colon cancer (OR, 1.50; 95% CI, 1.14-1.94; P = .0036). Prostate cancer was increased in first- (OR, 1.64; 95% CI, 1.18-2.23; P = .0026), second- (OR, 1.54; 95% CI, 1.32-1.79; P < .001), and third-degree relatives (OR, 1.33; 95% CI, 1.20-1.48; P < .001). CONCLUSION Data suggest common genetic risk for POI and reproductive cancers. Tools are needed to predict cancer risk in women with POI and potentially to counsel about risks of hormone replacement therapy.
Collapse
Affiliation(s)
- Kristina Allen-Brady
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Barry Moore
- Utah Center for Genetic Discovery, Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Lauren E Verrilli
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Intermountain Healthcare, Murray, UT 84107, USA
| | - Margaret A Alvord
- Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Marina Kern
- Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Nicola Camp
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Kristen Kelley
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Joseph Letourneau
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Lisa Cannon-Albright
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Mark Yandell
- Utah Center for Genetic Discovery, Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Erica B Johnstone
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Corrine K Welt
- Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
17
|
Decanter C, Dassonneville A, D'Orazio E, Behal H, Gagez AL, Mailliez A, Pigny P. AMH decline during chemotherapy reflects breast cancer cell DNA damage response (DDR) proficiency: the ONCO AMH1 pilot study. J Assist Reprod Genet 2025:10.1007/s10815-025-03475-9. [PMID: 40220108 DOI: 10.1007/s10815-025-03475-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
PURPOSE The impact of a germline BRCA1/2 pathogenic variant (gBRCApv) on baseline or late post-treatment AMH concentrations in breast cancer patients has been extensively studied, yielding mixed conclusions. However, whether the AMH decline during neo-adjuvant chemotherapy reflects differences in chemotherapy susceptibility between gBRCApv carriers and non-carriers remains unexplored. METHODS A monocentric, retrospective, longitudinal study was conducted on breast cancer patients carrying a gBRCApv (n = 12) or wild-type (WT) (n = 35) who received a neo-adjuvant sequential chemotherapy (CT) with anthracyclines followed by taxanes. Serum AMH levels were measured at baseline (AMH0) and at three time points during CT by a hypersensitive assay. Tumor size change was assessed via imaging. The impact of genetic status on AMH decline was evaluated using a linear mixed model with post hoc analysis. RESULTS The change of AMH concentrations from baseline to the end of CT tended to be influenced by the genetic status (BRCA * time interaction, p = 0.058). The slope between AMH0 and the end of anthracyclines (after log transformation) was steeper in gBRCApv than in WT patients (mean (SE): - 5.54 (0.63) vs - 3.97 (0.62); p = 0.023). Tumor size change was positively and significantly correlated with the change in AMH levels (AMH MidCT-AMH0) in gBRCApv patients (r = 0.93, p < 0.001) but not in WT patients (r = - 0.05; p = 0.84). CONCLUSION Germline BRCA1/2 status influences AMH decline during neo-adjuvant CT with drugs inducing DNA lesions. AMH decay is positively related to tumor size change assessed by imaging in gBRCApv patients. However, no conclusions can be drawn regarding the relationship with treatment response assessed by histological criteria.
Collapse
Affiliation(s)
- Christine Decanter
- Service d'Assistance Médicale à la Procréation et de Préservation de Fertilité, CHU Lille, 59037, Lille, France
- Univ. Lille, CANTHER, Inserm UMR 1277, 59045, Lille, France
| | - Audrey Dassonneville
- Service de Biochimie Hormonologie-Métabolisme-Nutrition-Oncologie, CHU Lille, 59037, Lille, France
| | - Emmanuelle D'Orazio
- Service d'Assistance Médicale à la Procréation et de Préservation de Fertilité, CHU Lille, 59037, Lille, France
| | - Hélène Behal
- Service de Biostatistiques, CHU Lille, 59000, Lille, France
| | - Anne-Laure Gagez
- Cellule Promotion, DRCI, Centre Oscar Lambret, BRT, 59020, Lille, France
| | - Audrey Mailliez
- Comité de Sénologie, Centre Oscar Lambret, Pôle d'Oncologie Médicale, 59020, Lille, France
| | - Pascal Pigny
- Service de Biochimie Hormonologie-Métabolisme-Nutrition-Oncologie, CHU Lille, 59037, Lille, France.
| |
Collapse
|
18
|
Carioscia SA, Biddanda A, Starostik MR, Tang X, Hoffmann ER, Demko ZP, McCoy RC. Common variation in meiosis genes shapes human recombination phenotypes and aneuploidy risk. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.02.25325097. [PMID: 40321295 PMCID: PMC12047964 DOI: 10.1101/2025.04.02.25325097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
The leading cause of human pregnancy loss is aneuploidy, often tracing to errors in chromosome segregation during female meiosis. While abnormal crossover recombination is known to confer risk for aneuploidy, limited data have hindered understanding of the potential shared genetic basis of these key molecular phenotypes. To address this gap, we performed retrospective analysis of preimplantation genetic testing data from 139,416 in vitro fertilized embryos from 22,850 sets of biological parents. By tracing transmission of haplotypes, we identified 3,656,198 crossovers, as well as 92,485 aneuploid chromosomes. Counts of crossovers were lower in aneuploid versus euploid embryos, consistent with their role in chromosome pairing and segregation. Our analyses further revealed that a common haplotype spanning the meiotic cohesin SMC1B is significantly associated with both crossover count and maternal meiotic aneuploidy, with evidence supporting a non-coding cis-regulatory mechanism. Transcriptome- and phenome-wide association tests also implicated variation in the synaptonemal complex component C14orf39 and crossover-regulating ubiquitin ligases CCNB1IP1 and RNF212 in meiotic aneuploidy risk. More broadly, recombination and aneuploidy possess a partially shared genetic basis that also overlaps with reproductive aging traits. Our findings highlight the dual role of recombination in generating genetic diversity, while ensuring meiotic fidelity.
Collapse
Affiliation(s)
| | - Arjun Biddanda
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Xiaona Tang
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Eva R. Hoffmann
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Rajiv C. McCoy
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
19
|
Cieri F, Cross CL, Francesco GD, Caldwell JZK. Dynamic neurocognitive adaptation: A follow-up exposome investigation in aging. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2025; 11:e70103. [PMID: 40390822 PMCID: PMC12086992 DOI: 10.1002/trc2.70103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 05/21/2025]
Abstract
INTRODUCTION Forty-five percent of Alzheimer's disease (AD) cases may be preventable. Validated tools for measuring environmental factors, with precision equal to that of current biological and genetic assessment tools, are currently lacking. METHODS We used the dynamic Neurocognitive Adaptation (dNA) scale, our validated tool to explore protective factors in AD, in 410 older adult participants (50% women). The dNA asks participants to recall cognitive, creative, physical, and social activities that they engaged in at seven different time periods in their lives. We examined associations among engagement in these domains using distance correlations and tested differences in domain engagement over time with repeated-measures analysis of variance. We calculated within-subjects comparisons for time and all interactions among time, sex, and education. We examined between-subjects factors for sex, education, and their interaction. From these models, we constructed visualizations of estimated marginal means against time to assess potential patterns of interest. RESULTS Physical and creative domain engagements were significantly correlated (p < 0.001) in the full sample, and social engagement correlated with physical (p < 0.001) and creative (p = 0.047) domains among females. Cognitive engagement increased over time (p < 0.001) for the full sample, while physical and creative engagement increased from childhood to adolescence, then decreased over time (p < 0.001). In contrast, social engagement increased from childhood to adolescence, declined through the senior years, and then sharply increased in old age. Overall, women showed higher cognitive engagement (p = 0.024) and men showed higher physical engagement (p = 0.011). Education was positively related to higher scores in all domains. DISCUSSION Our scale provides new insight into the correlation of environmental factors with education, suggests areas for lifestyle intervention, and highlights the importance of sex differences and middle age as a potential transition stage. Highlights Physical activity decreases and cognitive activity increases through time.Higher involvement in physical activities is correlated with creative and social dimensions.Men are more involved in physical and women in cognitive activities.Higher education is associated with higher involvement in all the dimensions explored.
Collapse
Affiliation(s)
- Filippo Cieri
- Department of NeurologyCleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
| | - Chad Lee Cross
- Department of NeurologyCleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
- Department of Epidemiology & Biostatistics (C.L.C.)School of Public HealthUniversity of NevadaLas VegasNevadaUSA
| | - Giulia Di Francesco
- Hematology UnitDepartment of Oncology‐HematologyPescara HospitalPescaraItaly
| | | |
Collapse
|
20
|
Liao CC, Lee CI, Li JM. Causal associations between dietary habits and age at natural menopause: A two-sample diet-wide Mendelian randomization study. Eur J Obstet Gynecol Reprod Biol 2025; 307:84-90. [PMID: 39893789 DOI: 10.1016/j.ejogrb.2025.01.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Menopause, the natural cessation of menstrual cycles, marks a significant phase in a woman's life and is associated with various health outcomes. While previous observational studies have suggested associations between dietary habits and the age at natural menopause (ANM), their findings have been inconsistent and often confounded, making it difficult to infer causality. This study aims to assess the causal relationships between dietary habits and ANM using a two-sample diet-wide Mendelian randomization (MR) approach. METHODS The study utilized data from 445,779 participants in the UK Biobank and genetic data from 201,323 women in the ReproGen Consortium. We selected single nucleotide polymorphisms associated with 38 dietary habits as instrumental variables for MR analysis. We applied MR methods, including inverse variance weighted (IVW), MR-Egger, weighted median, and weighted mode, to assess associations. Sensitivity analyses, including heterogeneity tests, pleiotropy assessments, and leave-one-out analyses, were conducted to validate the robustness of the findings. RESULTS Beer or cider consumption and spread on bread consumption were linked to an earlier onset of menopause. Oily fish consumption showed a potential association with a later onset of menopause, though this was not significant after false discovery rate correction. Sensitivity analyses confirmed the robustness of these findings, indicating no significant pleiotropy and stable effect estimates. CONCLUSION This study provides evidence that certain dietary habits may influence the timing of menopause. These insights underscore the importance of diet in reproductive aging and warrant further investigation to better understand these relationships and their underlying mechanisms.
Collapse
Affiliation(s)
- Chung-Chih Liao
- Department of Integrated Chinese and Western Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan; Department of Post-Baccalaureate Veterinary Medicine, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan
| | - Chun-I Lee
- Department of Obstetrics and Gynecology, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; Division of Infertility, Lee Women's Hospital, Taichung 40652, Taiwan; Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
| | - Jung-Miao Li
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan.
| |
Collapse
|
21
|
Dong J, Zou Z, Wang W, Chen L, Ma R, Ge X, Jing J, Ma J, Yao B. A novel homozygous frameshift mutation in MCM8 causes primary gonadal dysgenesis in both genders. J Assist Reprod Genet 2025; 42:1197-1204. [PMID: 40064807 PMCID: PMC12055739 DOI: 10.1007/s10815-025-03443-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/26/2025] [Indexed: 05/07/2025] Open
Abstract
PURPOSE We aimed to demonstrate the genetic factors of primary gonadal dysgenesis in a consanguineous family characterized by underdeveloped testes and non-obstructive azoospermia (NOA) in a male and primary amenorrhoea and primary ovarian insufficiency (POI) in a female. METHODS DNA was extracted from the male proband with infertility from the consanguineous family for whole-exome sequencing and bioinformatics analysis to screen for potential pathogenic genes and mutations. Sanger sequencing was used for further validation of his family pedigree. The effects of the identified novel mutation were evaluated in the male testes tissue by immunohistochemistry and in HEK293T cells by Western blot. RESULTS A homozygous frameshift mutation c.998delG (p. Gly333Glufs*50) in MCM8 was identified in the two siblings. The testes tissue sections in the male showed a Sertoli cell-only syndrome (SCOS). Functional analysis in vitro suggested that the mutation results in a truncated protein of MCM8 in HEK293T cells, and immunohistochemistry in vivo showed decreased expression of MCM8 protein. CONCLUSION We identified a novel homozygous frameshift mutation of MCM8 in two siblings diagnosed with primary gonadal dysgenesis from a consanguineous family. Functional analysis confirmed the pathogenicity of this mutation. Our study not only further reveals the essential role of MCM8 in human gonadal development, but also expands the mutational spectrum of MCM8 involved in male NOA and female POI and provides a new molecular marker for genetic counseling of infertility.
Collapse
Affiliation(s)
- Jie Dong
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Zhichuan Zou
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Wenhua Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Li Chen
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
- Department of Reproductive Medicine, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, 210002, Jiangsu, China
| | - Rujun Ma
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
- Department of Reproductive Medicine, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, 210002, Jiangsu, China
| | - Xie Ge
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
- Department of Reproductive Medicine, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, 210002, Jiangsu, China
| | - Jun Jing
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China
- Department of Reproductive Medicine, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, 210002, Jiangsu, China
| | - Jinzhao Ma
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China.
- Department of Reproductive Medicine, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, 210002, Jiangsu, China.
| | - Bing Yao
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, Jiangsu, China.
- Department of Reproductive Medicine, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, 210002, Jiangsu, China.
| |
Collapse
|
22
|
Zeng L, Liang Y, Huang L, Li Z, Kumar M, Zheng X, Li J, Luo S, Zhu L. Untargeted metabolomics reveals homogeneity and heterogeneity between physiological and pathological ovarian aging. J Ovarian Res 2025; 18:56. [PMID: 40098062 PMCID: PMC11912745 DOI: 10.1186/s13048-025-01625-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Ovarian aging is the main cause of reduced reproductive life span, yet its metabolic profiles remain poorly understood. This study aimed to reveal the metabolic homogeneity and heterogeneity between physiological and pathological ovarian aging. METHODS Seventy serum samples from physiological ovarian aging participants, pathological ovarian aging participants (including diminished ovarian reserve (DOR), subclinical premature ovarian insufficiency (scPOI) and premature ovarian insufficiency (POI)), as well as healthy participants were collected and analyzed by untargeted metabolomics. RESULTS Five homogeneous differential metabolites (neopterin, menaquinone, sphingomyelin (SM) (d14:1/24:2), SM (d14:0/21:1) and SM (d17:0/25:1)) were found in both physiological and pathological ovarian aging. While five distinct metabolites, including phosphoglyceride (PC) (17:0/18:2), PC (18:2e/17:2), SM (d22:1/14:1), SM (d14:1/20:1) and 4-hydroxyretinoic acid were specific to pathological ovarian aging. Functional annotation of differential metabolites suggested that folate biosynthesis, ubiquinone and other terpenoid-quinone biosynthesis pathways, were mainly involved in the ovarian aging process. Meanwhile, dopaminergic synapses pathway was strongly associated with scPOI, vitamin digestion and absorption and retinol metabolism were associated with POI. Furthermore, testosterone sulfate, SM (d14:0/28:1), PC (18:0e/4:0) and 4-hydroxyretinoic acid, were identified as potential biomarkers for diagnosing physiological ovarian aging, DOR, scPOI, and POI, respectively. Additionally, SM (d14:1/24:2) strongly correlated with both physiological and pathological ovarian aging. 4-hydroxyretinoic acid was strongly correlated with pathological ovarian aging. CONCLUSIONS Metabolic homogeneity of physiological and pathological ovarian aging was related to disorders of lipid, folate, ubiquinone metabolism, while metabolic heterogeneity between them was related to disorders of lipid, vitamin and retinol metabolism. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Lihua Zeng
- Department of Gynecology, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, Guangzhou, 514056, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yunyi Liang
- Department of Gynecology, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, Guangzhou, 514056, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Lizhi Huang
- Department of Gynecology, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, Guangzhou, 514056, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zu'ang Li
- Department of Gynecology, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, Guangzhou, 514056, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Manish Kumar
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiasheng Zheng
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jing Li
- Department of Gynecology, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, Guangzhou, 514056, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Songping Luo
- Department of Gynecology, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, Guangzhou, 514056, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Ling Zhu
- Department of Gynecology, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Lingnan Medical Research Center of Guangzhou, University of Chinese Medicine, Guangzhou, 514056, China.
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
23
|
Baker JL, Gordon-Dseagu VL, Voortman T, Chan D, Herceg Z, Robinson S, Norat T, Croker H, Ong K, Kampman E. Lifecourse research in cancer: context, challenges, and opportunities when exploring exposures in early life and cancer risk in adulthood. HEALTH OPEN RESEARCH 2025; 6:16. [PMID: 39974286 PMCID: PMC11836561 DOI: 10.12688/healthopenres.13748.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/12/2025] [Indexed: 02/21/2025]
Abstract
As the global population ages, and rates of modifiable risk factors for cancer change, cancer incidence and mortality continue to increase. While we understand many modifiable risk factors related to diet, nutrition, bodyweight, and physical activity in adulthood that influence cancer risk, how exposure during childhood, adolescence, and young adulthood impacts cancer risk is less clear. This is partly because the timeline from initial mutation to cancer development and diagnosis can span several decades. This long latency period creates methodological, ethical, and financial issues; as well as resource and feasibility challenges in the design, implementation, and data analysis of lifecourse studies. As such, the large majority of lifecourse studies are observational, often using recall data which has inherent bias issues. Concurrently, a new research era has begun, with mature birth cohort studies that are phenotyped/genotyped and can support studies on adult cancer risk. Several studies and consortia contain information spanning the lifecourse. These resources can support association, mechanistic and epigenetic investigations into the influences of multi-disciplinary (e.g. genetic, behavioural, environmental) factors, across the lifecourse and critical time periods. Ultimately, we will be able to produce high-quality evidence and identify how/when early life risk factors impact cancer development and survival.
Collapse
Affiliation(s)
- Jennifer L. Baker
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, University of Copenhagen, Frederiksberg, Denmark
| | | | - Trudy Voortman
- Department of Epidemiology, Erasmus MC, Erasmus, Rotterdam, The Netherlands
| | - Doris Chan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, England, UK
| | - Zdenko Herceg
- International Agency for Research on Cancer (IARC), World Health Organisation, Lyon, France
| | - Sian Robinson
- AGE Research Group, Newcastle University, Newcastle upon Tyne, England, UK
| | - Teresa Norat
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, England, UK
| | - Helen Croker
- World Cancer Research Fund International, London, England, UK
| | - Ken Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, England, UK
| | - Ellen Kampman
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
24
|
Wu D, Liu C, Ding L. Follicular metabolic dysfunction, oocyte aneuploidy and ovarian aging: a review. J Ovarian Res 2025; 18:53. [PMID: 40075456 PMCID: PMC11900476 DOI: 10.1186/s13048-025-01633-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
With the development of modern society and prolonged education, more women choose to delay their childbearing age, which greatly increases the number of women aged older than 35 years with childbearing needs. However, with increasing age, the quantity and quality of oocytes continue to fall, especially with increasing aneuploidy, which leads to a low in vitro fertilization (IVF) success rate, high abortion rate and high teratogenesis rate in assisted reproduction in women with advanced maternal age. In addition to genetics and epigenetics, follicular metabolism homeostasis is closely related to ovarian aging and oocyte aneuploidy. Glucose, lipid, and amino acid metabolism not only provide energy for follicle genesis but also regulate oocyte development and maturation. This review focuses on the relationships among follicular metabolism, oocyte aneuploidy, and ovarian aging and discusses potential therapeutic metabolites for ovarian aging.
Collapse
Affiliation(s)
- Die Wu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, 210008, China
| | - Chuanming Liu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, 210008, China
| | - Lijun Ding
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China.
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, 210008, China.
- State Key Laboratory of Analytic Chemistry for Life Science, Nanjing University, Nanjing, 210093, China.
- Clinical Center for Stem Cell Research, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
25
|
Pujol Gualdo N, Džigurski J, Rukins V, Pajuste FD, Wolford BN, Võsa M, Golob M, Haug L, Alver M, Läll K, Peters M, Brumpton BM, Palta P, Mägi R, Laisk T. Atlas of genetic and phenotypic associations across 42 female reproductive health diagnoses. Nat Med 2025:10.1038/s41591-025-03543-8. [PMID: 40069456 DOI: 10.1038/s41591-025-03543-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/28/2025] [Indexed: 04/02/2025]
Abstract
The genetic background of many female reproductive health diagnoses remains uncharacterized, compromising our understanding of the underlying biology. Here, we map the genetic architecture across 42 female-specific health conditions using data from up to 293,618 women from two large population-based cohorts, the Estonian Biobank and the FinnGen study. Our study illustrates the utility of genetic analyses in understanding women's health better. As specific examples, we describe genetic risk factors for ovarian cysts that elucidate the genetic determinants of folliculogenesis and, by leveraging population-specific variants, uncover new candidate genes for uterine fibroids. We find that most female reproductive health diagnoses have a heritable component, with varying degrees of polygenicity and discoverability. Finally, we identify pleiotropic loci and genes that function in genital tract development (WNT4, PAX8, WT1, SALL1), hormonal regulation (FSHB, GREB1, BMPR1B, SYNE1/ESR1) and folliculogenesis (CHEK2), underlining their integral roles in female reproductive health.
Collapse
Affiliation(s)
- Natàlia Pujol Gualdo
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jelisaveta Džigurski
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Valentina Rukins
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Fanny-Dhelia Pajuste
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Brooke N Wolford
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mariann Võsa
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mia Golob
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Lisette Haug
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Maris Alver
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kristi Läll
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Maire Peters
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Celvia CC AS, Tartu, Estonia
| | - Ben M Brumpton
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Priit Palta
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Triin Laisk
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia.
| |
Collapse
|
26
|
Kuchel GA, Hevener AL, Ruby JG, Sebastiani P, Kumar V. Workshop Report-Heterogeneity and Successful Aging Part I: Heterogeneity in Aging-Challenges and Opportunities. J Gerontol A Biol Sci Med Sci 2025; 80:glaf023. [PMID: 40052564 DOI: 10.1093/gerona/glaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Indexed: 05/13/2025] Open
Abstract
Historically, aging research has focused primarily on the study of differences in means of varied measures obtained at different ages. However, growing evidence has shown that for many parameters, variability in measurements obtained both between- and within-age groups increases with aging. Moreover, growing heterogeneity may become especially apparent when examined via longitudinal as opposed to cross-sectional aging data. Efforts to deconvolute and better understand such heterogeneity present remarkable translational opportunities for developing targeted and more effective interventions into aging. Here, we present Part I, a summary of the NIA Heterogeneity and Successful Aging workshop virtually held in May 2023.
Collapse
Affiliation(s)
- George A Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Andrea L Hevener
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - J Graham Ruby
- Calico Life Sciences LLC, South San Francisco, California, USA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Vivek Kumar
- The Jackson Laboratory, Bar Harbor, Maine, USA
| |
Collapse
|
27
|
Dicks EM, Tyrer JP, Ezquina S, Jones M, Baierl J, Peng PC, Diaz M, Goode E, Winham SJ, Dörk T, Gorp TV, Fazio AD, Bowtell DDL, Garsed DW, Odunsi K, Moysich K, Pavanello M, Fostira F, Webb PM, Soukupová J, Cohen PA, Sieh W, Fortner RT, Ricker C, Karlan B, Campbell I, Brenton JD, Ramus SJ, Gayther SA, Pharoah PDP. Exome sequencing identifies HELB as a novel susceptibility gene for non-mucinous, non-high-grade-serous epithelial ovarian cancer. Eur J Hum Genet 2025; 33:297-303. [PMID: 39939714 PMCID: PMC11894177 DOI: 10.1038/s41431-025-01786-0] [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: 10/04/2024] [Revised: 12/23/2024] [Accepted: 01/09/2025] [Indexed: 02/14/2025] Open
Abstract
Rare, germline loss-of-function variants in a handful of DNA repair genes are associated with epithelial ovarian cancer. The aim of this study was to evaluate the role of rare, coding, loss-of-function variants across the genome in epithelial ovarian cancer. We carried out a gene-by-gene burden test with various histotypes using data from 2573 non-mucinous cases and 13,923 controls. Twelve genes were associated at a False Discovery Rate of less than 0.1 of which seven were the known ovarian cancer susceptibility genes BRCA1, BRCA2, BRIP1, RAD51C, RAD51D, MSH6 and PALB2. The other five genes were OR2T35, HELB, MYO1A and GABRP which were associated with non-high-grade serous ovarian cancer and MIGA1 which was associated with high-grade serous ovarian cancer. Further support for the association of HELB association comes from the observation that loss-of-function variants in HELB are associated with age at natural menopause and Mendelian randomisation analysis shows an association between genetically predicted age at natural menopause and endometrioid ovarian cancer, but not high-grade serous ovarian cancer.
Collapse
Affiliation(s)
- Ed M Dicks
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Jonthan P Tyrer
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Suzana Ezquina
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Michelle Jones
- Department of Biomedical Sciences, Cedars-Sinai Medical Centre, Los Angeles, CA, USA
| | - John Baierl
- Department of Computational Biomedicine, Cedars-Sinai Medical Centre, Los Angeles, CA, USA
| | - Pei-Chen Peng
- Department of Computational Biomedicine, Cedars-Sinai Medical Centre, Los Angeles, CA, USA
| | - Michael Diaz
- Department of Biomedical Sciences, Cedars-Sinai Medical Centre, Los Angeles, CA, USA
| | | | | | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Toon Van Gorp
- Division of Gynaecological Oncology, Leuven Cancer Institute, University Hospital Leuven and KU Leuven, Leuven, Belgium
| | - Anna De Fazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- The Daffodil Centre, The University of Sydney, A JOINT Venture with Cancer Council NSW, Sydney, NSW, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
| | - David D L Bowtell
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Dale W Garsed
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Kunle Odunsi
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, USA
| | - Kirsten Moysich
- Division of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Marina Pavanello
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia
| | - Florentia Fostira
- Human Molecular Genetics Laboratory, National Centre for Scientific Research, Athens, Greece
| | - Penelope M Webb
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jana Soukupová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Paul A Cohen
- Division of Obstetrics and Gynaecology, Medical School, University of Western Australia, Crawley, WA, Australia
| | - Weiva Sieh
- MD Anderson Cancer Center, Houston, TX, USA
| | - Renée Turzanski Fortner
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Charite Ricker
- Keck School of Medicine, Division of Medical Oncology, University of Southern California, Los Angeles, CA, USA
| | - Beth Karlan
- University of California Los Angeles, Los Angeles, CA, USA
| | - Ian Campbell
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - James D Brenton
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Susan J Ramus
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, NSW, Australia
- Adult Cancer Program, Lowy Cancer Research Centre, University of NSW, Sydney, NSW, Australia
| | - Simon A Gayther
- Center for Inherited Oncogenesis, Department of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| | - Paul D P Pharoah
- Department of Computational Biomedicine, Cedars-Sinai Medical Centre, Los Angeles, CA, USA.
| |
Collapse
|
28
|
Wang X, Dossus L, Gunter MJ, Crosbie EJ, Ong JS, Glubb DM, O'Mara TA. Risk Stratification for Endometrial Cancer Reveals Independent Contributions of Polygenic Risk and Body Mass Index. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.19.25322538. [PMID: 40034786 PMCID: PMC11875273 DOI: 10.1101/2025.02.19.25322538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background Obesity is a major risk factor for endometrial cancer, but it is unknown whether it impacts the association between genetic risk and endometrial cancer. We incorporated polygenic risk score and epidemiological risk factors in the prediction of and investigated associations of BMI and polygenic risk score with endometrial cancer risk. Methods We generated polygenic risk score for endometrial cancer in 129,829 unrelated female participants of European ancestry (including 956 incident cases with endometrial cancer) in the UK Biobank and predicted endometrial cancer using endometrial cancer polygenic risk score and established epidemiological risk factors, including BMI. We evaluated the performance of endometrial cancer prediction models by odds ratios and area under the receiver operating characteristic curves (AUCs) to using logistic regression. Individual and joint associations of BMI and polygenic risk score with endometrial cancer were assessed using Cox proportional hazards models. Results An integrated model incorporating both polygenic risk score and epidemiological risk factors achieved a modest, but statistically significant, improvement in predicting endometrial cancer status compared with the model that included epidemiologic risk factors alone (AUC = 0.74 versus 0.73; P = 3.98 × 10 -5 ). Obese participants (BMI ≥ 30 kg/m 2 ) in the top polygenic risk tertile had the highest endometrial cancer risk. We observed independent effects of genetic risk and BMI on endometrial cancer risk. Conclusion Integrating polygenic risk score with epidemiological risk factors may offer insights into population stratification for endometrial cancer susceptibility. Higher endometrial cancer polygenic risk is associated with endometrial cancer, irrespective of BMI.
Collapse
|
29
|
Santoro N. Understanding the menopause journey. Climacteric 2025:1-5. [PMID: 39903209 DOI: 10.1080/13697137.2024.2445303] [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: 11/08/2024] [Revised: 12/01/2024] [Accepted: 12/10/2024] [Indexed: 02/06/2025]
Abstract
The menopause experience is unique to people with ovaries who attain an age at which functioning follicles are depleted. Unlike male reproductive aging, menopause is accompanied by a definable reproductive milestone in that menstrual periods cease and the failure of follicle growth results in a large drop in circulating estrogen and no further ovarian production of progesterone. While the focus on menopause has largely been centered on this absence of hormone production, the most dynamic changes in symptoms and health markers begin before the final menstrual period, and merit attention. Vasomotor symptoms, the most common symptom of menopause and the primary symptom that drives women to seek treatment, peak in frequency and prevalence in the late menopause transition, when women are still having menstrual periods. Body composition and adverse lipoprotein and lipid changes also worsen most acutely in the late transition, and then assume a slower, age-related trajectory of change. Multiple processes that worsen across the transition restabilize after it is over. The notion that the menopause transition is an adaptive process for women has scientific merit and suggests that facilitating this adaptation and recognizing its implications may represent the next phase of progress in the field.
Collapse
Affiliation(s)
- Nanette Santoro
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
30
|
Tang W, Wang K, Feng Y, Tsui KH, Singh KK, Stout MB, Wang S, Wu M. Exploration of the mechanism and therapy of ovarian aging by targeting cellular senescence. LIFE MEDICINE 2025; 4:lnaf004. [PMID: 40110109 PMCID: PMC11916902 DOI: 10.1093/lifemedi/lnaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/22/2025] [Indexed: 03/22/2025]
Abstract
The ovary is a crucial gonadal organ that supports female reproductive and endocrine functions. Ovarian aging can result in decreased fertility and dysfunction across multiple organs. Research has demonstrated that cellular senescence in various cell types within the ovary can trigger a decline in ovarian function through distinct stress responses, resulting in ovarian aging. This review explores how cellular senescence may contribute to ovarian aging and reproductive failure. Additionally, we discuss the factors that cause ovarian cellular senescence, including the accumulation of advanced glycation end products, oxidative stress, mitochondrial dysfunction, DNA damage, telomere shortening, and exposure to chemotherapy. Furthermore, we discuss senescence in six distinct cell types, including oocytes, granulosa cells, ovarian theca cells, immune cells, ovarian surface epithelium, and ovarian endothelial cells, inside the ovary and explore their contribution to the accelerated ovarian aging. Lastly, we describe potential senotherapeutics for the treatment of ovarian aging and offer novel strategies for ovarian longevity.
Collapse
Affiliation(s)
- Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| | - Kaichen Wang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yourong Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813779, Taiwan, China
- Department of Obstetrics and Gynecology, Yang-Ming University, Taipei 112304, Taiwan, China
- Department of Pharmacy and Graduate Institute of Pharmaceutical Technology, Tajen University, Pingtung 900391, Taiwan, China
| | - Keshav K Singh
- Department of Genetics, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| |
Collapse
|
31
|
Multiomics atlas reveals molecular and genetic drivers of human ovarian aging. NATURE AGING 2025; 5:186-187. [PMID: 39910245 DOI: 10.1038/s43587-025-00821-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
|
32
|
Jin C, Wang X, Yang J, Kim S, Hudgins AD, Gamliel A, Pei M, Contreras D, Devos M, Guo Q, Vijg J, Conti M, Hoeijmakers J, Campisi J, Lobo R, Williams Z, Rosenfeld MG, Suh Y. Molecular and genetic insights into human ovarian aging from single-nuclei multi-omics analyses. NATURE AGING 2025; 5:275-290. [PMID: 39578560 PMCID: PMC11839473 DOI: 10.1038/s43587-024-00762-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
The ovary is the first organ to age in the human body, affecting both fertility and overall health. However, the biological mechanisms underlying human ovarian aging remain poorly understood. Here we present a comprehensive single-nuclei multi-omics atlas of four young (ages 23-29 years) and four reproductively aged (ages 49-54 years) human ovaries. Our analyses reveal coordinated changes in transcriptomes and chromatin accessibilities across cell types in the ovary during aging, notably mTOR signaling being a prominent ovary-specific aging pathway. Cell-type-specific regulatory networks reveal enhanced activity of the transcription factor CEBPD across cell types in the aged ovary. Integration of our multi-omics data with genetic variants associated with age at natural menopause demonstrates a global impact of functional variants on gene regulatory networks across ovarian cell types. We nominate functional non-coding regulatory variants, their target genes and ovarian cell types and regulatory mechanisms. This atlas provides a valuable resource for understanding the cellular, molecular and genetic basis of human ovarian aging.
Collapse
Affiliation(s)
- Chen Jin
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Xizhe Wang
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jiping Yang
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Seungsoo Kim
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Adam D Hudgins
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Amir Gamliel
- Howard Hughes Medical Institute, Department and School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Mingzhuo Pei
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Daniela Contreras
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Melody Devos
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Qinghua Guo
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Marco Conti
- Center for Reproductive Sciences, University of California, San Francico, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jan Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Oncode Institute, Utrecht, The Netherlands
- Institute for Genome Stability in Ageing and Disease, Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University Hospital of Cologne, Cologne, Germany
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Rogerio Lobo
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Zev Williams
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute, Department and School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
33
|
Munakata Y, Hu M, Kitamura Y, Dani R, Bynder A, Fritz A, Schultz R, Namekawa S. Chromatin remodeler CHD4 establishes chromatin states required for ovarian reserve formation, maintenance and male germ cell survival. Nucleic Acids Res 2025; 53:gkaf008. [PMID: 39907108 PMCID: PMC11795200 DOI: 10.1093/nar/gkaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/13/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025] Open
Abstract
The ovarian reserve defines female reproductive lifespan, which in humans spans decades due to the maintenance of meiotic arrest in non-growing oocytes (NGOs) residing in primordial follicles. Unknown is how the chromatin state of NGOs is established to enable long-term maintenance of the ovarian reserve. Here, we show that a chromatin remodeler, CHD4, a member of the Nucleosome Remodeling and Deacetylase (NuRD) complex, establishes chromatin states required for formation and maintenance of the ovarian reserve. Conditional loss of CHD4 in perinatal mouse oocytes results in acute death of NGOs and depletion of the ovarian reserve. CHD4 establishes closed chromatin at regulatory elements of pro-apoptotic genes to prevent cell death and at specific genes required for meiotic prophase I to facilitate the transition from meiotic prophase I oocytes to meiotically-arrested NGOs. In male germ cells, CHD4 establishes closed chromatin at the regulatory elements of pro-apoptotic genes, allowing germ cell survival. These results demonstrate a role for CHD4 in defining a chromatin state that ensures germ cell survival, thereby enabling the long-term maintenance of both female and male germ cells.
Collapse
Affiliation(s)
- Yasuhisa Munakata
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA
| | - Mengwen Hu
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA
| | - Yuka Kitamura
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA
| | - Raissa G Dani
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA
| | - Adam L Bynder
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA
| | - Amelia S Fritz
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA
| | - Richard M Schultz
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Satoshi H Namekawa
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA
| |
Collapse
|
34
|
Li Z, Lin Y, Zou Y, Liang Y, Zeng L, Wang Y, Li Y, Zong Y, Zhang Y, Zheng Y, Cui Y, Huang L, Chen Z, Pan X, Zhu L. Zuogui pills ameliorate chemotherapy-induced ovarian aging by improving stemness, regulating cell cycle and reducing apoptosis of oogonial stem cells via the Notch1/Nrf2 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119105. [PMID: 39580130 DOI: 10.1016/j.jep.2024.119105] [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: 10/03/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zuogui Pills (ZGP) is a classic traditional Chinese herbal formula originating from the Ming Dynasty. It has been widely used in the treatment of kidney deficiency-related diseases, including ovarian aging. AIM OF THE STUDY To investigate the effects and potential mechanisms of ZGP on ovarian aging induced by the chemotherapeutic agent cyclophosphamide (CTX), as well as its impact on the therapeutic target, oogonial stem cells (OSCs), involving the Notch1/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. MATERIALS AND METHODS This study utilized High-Performance Liquid Chromatography (HPLC) to analyze the active components of Zuogui Pills (ZGP). In vivo experiments involved the establishment of an ovarian aging model in female rats through intraperitoneal injection of CTX, followed by an 8-week treatment with ZGP and dehydroepiandrosterone (DHEA). The Notch pathway inhibitor DAPT was administered via intraperitoneal injection, followed by ZGP intervention to validate its therapeutic effects. Transcriptomic sequencing was used to analyze the differential genes before and after ZGP treatment of CTX-induced ovarian aging, and KEGG and GO analyses were applied to assess the changes in relevant signaling pathways and biological processes. In vitro experiments included the extraction, separation, and purification of ovarian germ stem cells, followed by transfection with a Notch1 overexpression plasmid. The CTX active component 4-Hydroxycyclophosphamide (4HC) was used for model intervention, and ZGP, DHEA-containing serum, and DAPT were applied to intervene with the oogonial stem cells. The effects of CTX modeling, the therapeutic efficacy of ZGP, and the general condition of the rats were observed. H&E staining was employed to assess ovarian morphology and follicle counting at various stages. Serum hormone levels were measured using ELISA, while qPCR, Western blot, flow cytometry, immunofluorescence, and IHC were utilized to analyze the expression of the Notch1/Nrf2 pathway, cell cycle proteins, and stemness-related indicators. Flow cytometry, TUNEL fluorescence, and CCK8 assays were conducted to evaluate changes in cell cycle composition, apoptosis, and proliferation. Finally, ChIP-qPCR was employed to validate the transcriptional regulation of the target gene NFE2L2 by Notch1. RESULTS ZGP improved serum sex hormones in ovarian aging rats, enhanced ovarian index, and optimized ovarian and uterine morphology, as well as follicle quantity composition. After transcriptome sequencing, KEGG analysis enriched the Notch signaling pathway and cell cycle, while GO analysis highlighted enrichment in the Notch pathway and stem cell population maintenance. Various experiments validated that ZGP significantly improved the expression of cell cycle-related proteins Cyclin D1 (CCND1), Cyclin E1 (CCNE1), cyclin-dependent kinase inhibitor 1a (CDKN1A), stemness markers Mouse Vasa Homolog (MVH), Octamer-binding Transcription Factor 4 (Oct4), Fragilis, 5-Bromo-2'-deoxyuridine (BrdU), as well as Notch1 and Nrf2 in aging ovarian tissues and OSCs. Additionally, ZGP promoted the proliferation of 4HC-damaged OSCs, optimized OSCs cell cycle composition, reduced G0/G1 phase arrest, and decreased early and late apoptosis. ZGP could reverse the detrimental effects on stemness and cell cycle of OSCs caused by blocking the Notch pathway. Furthermore, ZGP may activate the regulation of its target gene NFE2L2 by upregulating Notch1 expression in OSCs, thereby exerting therapeutic effects. CONCLUSION ZGP protects ovarian function in CTX-induced ovarian aging rats by regulating the Notch1/Nrf2 pathway. It restores serum sex hormone levels, maintains normal follicle development, promotes the proliferation of aged OSCs, optimizes the cell cycle, reduces apoptosis, and preserves stemness, thereby alleviating ovarian aging.
Collapse
Affiliation(s)
- 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.
| | - Yuxin Zou
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yunyi Liang
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Lihua Zeng
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yixuan Wang
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Lingnan Medical Research Center of 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.
| | - Yun Zong
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yuying Zhang
- 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.
| | - Yixuan Cui
- 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.
| | - Xinyi Pan
- 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
|
35
|
Abt KM, Bartholomew MA, Nixon A, Richman HE, Gura MA, Seymour KA, Freiman RN. Transcriptional Integration of Meiotic Prophase I Progression and Early Oocyte Differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631470. [PMID: 39829852 PMCID: PMC11741336 DOI: 10.1101/2025.01.06.631470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Female reproductive senescence results from the regulated depletion of a finite pool of oocytes called the ovarian reserve. This pool of oocytes is initially established during fetal development, but the oocytes that comprise it must remain quiescent for decades until they are activated during maturation in adulthood. In order for developmentally competent oocytes to populate the ovarian reserve they must successfully initiate both meiosis and oogenesis. As the factors that regulate the timing and fidelity of these early events remain elusive, we assessed the precise function and timing of the transcriptional regulator TAF4b during meiotic prophase I progression in mouse fetal oocytes. Compared to matched controls, E14.5 Taf4b-deficient oocytes enter meiosis I in a timely manner however, their subsequent progression through the pachytene-to-diplotene transition of meiotic prophase I is compromised. Moreover, this disruption of meiotic progression is associated with the reduced ability of Taf4b-deficient oocytes to repair double-strand DNA breaks. Transcriptional profiling of Taf4b-deficient oocytes reveals that between E16.5 and E18.5 these oocytes fail to coordinate the reduction of meiotic gene expression and the induction of oocyte differentiation genes. These studies reveal that TAF4b promotes the formation of the ovarian reserve in part by orchestrating the timely transition to meiosis I arrest and oocyte differentiation, which are often perceived as separate events.
Collapse
Affiliation(s)
- Kimberly M. Abt
- MCB Graduate Program, Cell Biology, and Biochemistry, Brown University, 70 Ship St., Box G-E4, Providence, RI 02903, USA
| | - Myles A. Bartholomew
- MCB Graduate Program, Cell Biology, and Biochemistry, Brown University, 70 Ship St., Box G-E4, Providence, RI 02903, USA
| | - Anna Nixon
- MCB Graduate Program, Cell Biology, and Biochemistry, Brown University, 70 Ship St., Box G-E4, Providence, RI 02903, USA
| | - Hanna E. Richman
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship St., Box G-E4, Providence, RI 02903, USA
| | - Megan A. Gura
- MCB Graduate Program, Cell Biology, and Biochemistry, Brown University, 70 Ship St., Box G-E4, Providence, RI 02903, USA
| | - Kimberly A. Seymour
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship St., Box G-E4, Providence, RI 02903, USA
| | - Richard N. Freiman
- MCB Graduate Program, Cell Biology, and Biochemistry, Brown University, 70 Ship St., Box G-E4, Providence, RI 02903, USA
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship St., Box G-E4, Providence, RI 02903, USA
| |
Collapse
|
36
|
Mills M, Emori C, Kumar P, Boucher Z, George J, Bolcun-Filas E. Single-cell and bulk transcriptional profiling of mouse ovaries reveals novel genes and pathways associated with DNA damage response in oocytes. Dev Biol 2025; 517:55-72. [PMID: 39306223 DOI: 10.1016/j.ydbio.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Immature oocytes enclosed in primordial follicles stored in female ovaries are under constant threat of DNA damage induced by endogenous and exogenous factors. Checkpoint kinase 2 (CHEK2) is a key mediator of the DNA damage response (DDR) in all cells. Genetic studies have shown that CHEK2 and its downstream targets, p53, and TAp63, regulate primordial follicle elimination in response to DNA damage. However, the mechanism leading to their demise is still poorly characterized. Single-cell and bulk RNA sequencing were used to determine the DDR in wild-type and Chek2-deficient ovaries. A low but oocyte-lethal dose of ionizing radiation induces ovarian DDR that is solely dependent on CHEK2. DNA damage activates multiple response pathways related to apoptosis, p53, interferon signaling, inflammation, cell adhesion, and intercellular communication. These pathways are differentially employed by different ovarian cell types, with oocytes disproportionately affected by radiation. Novel genes and pathways are induced by radiation specifically in oocytes, shedding light on their sensitivity to DNA damage, and implicating a coordinated response between oocytes and pregranulosa cells within the follicle. These findings provide a foundation for future studies on the specific mechanisms regulating oocyte survival in the context of aging, therapeutic and environmental genotoxic exposures.
Collapse
Affiliation(s)
- Monique Mills
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA; The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04469, USA
| | - Chihiro Emori
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 5650871, Japan
| | - Parveen Kumar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06110, USA
| | - Zachary Boucher
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Joshy George
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06110, USA
| | | |
Collapse
|
37
|
Ruotsalainen S, Karjalainen J, Kurki M, Lahtela E, Pirinen M, Riikonen J, Ritari J, Tammi S, Partanen J, Laivuori H, Palotie A, Heyne H, Daly M, Widen E. Inherited infertility: Mapping loci associated with impaired female reproduction. Am J Hum Genet 2024; 111:2789-2798. [PMID: 39566493 PMCID: PMC11639076 DOI: 10.1016/j.ajhg.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
Female infertility is a common and complex health problem affecting millions of women worldwide. While multiple factors can contribute to this condition, the underlying cause remains elusive in up to 15%-30% of affected individuals. In our large genome-wide association study (GWAS) of 22,849 women with infertility and 198,989 control individuals from the Finnish population cohort FinnGen, we unveil a landscape of genetic factors associated with the disorder. Our recessive analysis identified a low-frequency stop-gained mutation in TATA-box binding protein-like 2 (TBPL2; c.895A>T [p.Arg299Ter]; minor-allele frequency [MAF] = 1.2%) with an impact comparable to highly penetrant monogenic mutations (odds ratio [OR] = 650, p = 4.1 × 10-25). While previous studies have linked the orthologous gene to anovulation and sterility in knockout mice, the severe consequence of the p.Arg299Ter variant was evidenced by individuals carrying two copies of that variant having significantly fewer offspring (average of 0.16) compared to women belonging to the other genotype groups (average of 1.75 offspring, p = 1.4 × 10-15). Notably, all homozygous women who had given birth had received infertility therapy. Moreover, our age-stratified analyses identified three additional genome-wide significant loci. Two loci were associated with early-onset infertility (diagnosed before age 30), located near CHEK2 and within the major histocompatibility complex (MHC) region. The third locus, associated with late-onset infertility, had its lead SNP located in an intron of a long non-coding RNA (lncRNA) gene. Taken together, our data highlight the significance of rare recessive alleles in shaping female infertility risk. The results further provide evidence supporting specific age-dependent mechanisms underlying this complex disorder.
Collapse
Affiliation(s)
- Sanni Ruotsalainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Juha Karjalainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Program for Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mitja Kurki
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Program for Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Elisa Lahtela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Matti Pirinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Juha Riikonen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jarmo Ritari
- Finnish Red Cross Blood Service, R&D, Helsinki, Finland
| | - Silja Tammi
- Finnish Red Cross Blood Service, R&D, Helsinki, Finland
| | | | - Hannele Laivuori
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland; Faculty of Medicine and Health Technology, Center for Child, Adolescent and Maternal Health Research, University of Tampere, Tampere, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Program for Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Henrike Heyne
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Program for Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Digital Health Center, Hasso Plattner Institute for Digital Engineering, University of Potsdam, Potsdam, Germany; Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark Daly
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Program for Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
38
|
Kloster AK, Adami LNG, Moysés-Oliveira M, Hachul H, Andersen ML, Tufik S. Exploring the Link between Premature Ovarian Insufficiency, Insomnia and Circadian Pathways. JBRA Assist Reprod 2024; 28:567-589. [PMID: 39254468 PMCID: PMC11622404 DOI: 10.5935/1518-0557.20240052] [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: 01/05/2024] [Accepted: 07/25/2024] [Indexed: 09/11/2024] Open
Abstract
OBJECTIVE To establish an interaction network for genes related to premature ovarian insufficiency (POI) and insomnia, and to identify biological processes that connect POI to the physiological clock. METHODS Previously reported lists of genes associated to POI and insomnia were contrasted and their intersection was used as input on protein-protein interaction analyses. POI-associated genes were contrasted with gene expression markers for neural circadian control and enriched pathways among their shared content were dissected. RESULTS The functional network generated from the intersection between POI and insomnia gene lists pointed to the central nervous system as the most relevant cellular context for this connection. After identifying POI-associated genes that play a role in neural circadian patterns, we observed the disruption of pathways related to the hypothalamic-pituitary-gonadal axis as the major genetic link between ovarian function and circadian neural circuits. CONCLUSIONS These findings highlight neurological mechanisms that support the POI-insomnia interplay.
Collapse
Affiliation(s)
- Anna K. Kloster
- Sleep Institute, Associação Fundo de Incentivo
à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São
Paulo, São Paulo, Brazil
| | - Luana N. G. Adami
- Sleep Institute, Associação Fundo de Incentivo
à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São
Paulo, São Paulo, Brazil
| | | | - Helena Hachul
- Sleep Institute, Associação Fundo de Incentivo
à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São
Paulo, São Paulo, Brazil
| | - Monica L. Andersen
- Sleep Institute, Associação Fundo de Incentivo
à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São
Paulo, São Paulo, Brazil
| | - Sergio Tufik
- Sleep Institute, Associação Fundo de Incentivo
à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São
Paulo, São Paulo, Brazil
| |
Collapse
|
39
|
Zhong W, Wang Q, Peng D, Zou Y, Chen Y, Xia Y, Zhang X, Shu M, Song C, Wang Y, Fu Y, Wang S, Ma Y, Bu X, Liang Y, Chen Y, Bai W, Chen Y, Deng C, Zhang W, Zhou M, Lv L, Zhang L, Lu S, Shang W. Genetic risk stratification and risk factors of early menopause in women: a multi-center study utilizing polygenic risk scores. Front Endocrinol (Lausanne) 2024; 15:1518288. [PMID: 39687074 PMCID: PMC11646720 DOI: 10.3389/fendo.2024.1518288] [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: 10/28/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Objective This study aims to evaluate the utility of polygenic risk scores (PRS) in women with early menopause (EM) and to investigate the clinical characteristics and risk factors associated with EM based on genetic risk. Study design Genotyping data and clinical data from women with EM and women with normal age of menopause retrieved from UK Biobank were used for early menopause risk prediction model establishment. Subsequently, 99 women diagnosed with EM and 1027 control women underwent PGT-M were recruited for model validation from across eight hospitals in China. According to PRS percentiles, these participants were further classified into high risk and intermediate risk groups. Characteristics among women at different risk levels were compared, and risk factors with early menopause were also statistical analyzed. Main outcome measures The proportion of women at high risk in EM and control groups; Characteristics with significant difference among women at different risk levels; risk factors associated with EM. Results The proportion of high-risk women in the EM group was significantly higher than that in control women underwent PGT-M (Group PGT-M) (OR = 3.78), and that in women with normal age menopause from UK Biobank (Group UKB) (OR = 5.11). Notably, the women with high risk of EM exhibited distinct characteristics compared to women with the intermediate-risk of EM, and identified several risk factors associated with EM. Conclusions We established a PRS model to serves as a valuable instrument for EM risk prediction. The exploratory analysis revealed that women with high risk of EM exhibited a higher height, suggesting EM related genetic loci may also influence growth and development level. Several risk factors were found to be potentially associated with EM, such as excessive familial contentment, COVID-19 vaccination, staying up late, and the husband's engagement in smoking and alcohol abuse.
Collapse
Affiliation(s)
- Wei Zhong
- Department of Obstetrics and Gynecology, The Seventh Medical Center of People’s Liberation Army General Hospital, Beijing, China
- Department of Obstetrics and Gynecology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
- Department of Obstetrics and Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Qihang Wang
- Department of Obstetrics and Gynecology, The Seventh Medical Center of People’s Liberation Army General Hospital, Beijing, China
| | - Dingchuan Peng
- Department of Obstetrics and Gynecology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Yangyun Zou
- Department of Clinical Research, Yikon Genomics Co., Ltd, Suzhou, Jiangsu, China
| | - Yulin Chen
- Department of Clinical Research, Yikon Genomics Co., Ltd, Suzhou, Jiangsu, China
| | - Yingying Xia
- Department of Clinical Research, Yikon Genomics Co., Ltd, Suzhou, Jiangsu, China
| | - Xin Zhang
- Department of Clinical Research, Yikon Genomics Co., Ltd, Suzhou, Jiangsu, China
| | - Mingming Shu
- Department of Obstetrics and Gynecology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Chunlan Song
- Department of Obstetrics and Gynecology, The Seventh Medical Center of People’s Liberation Army General Hospital, Beijing, China
| | - Yiran Wang
- Department of Obstetrics and Gynecology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Yiyao Fu
- Department of Obstetrics and Gynecology, Chinese People's Liberation Army Medical School, Beijing, China
| | - Sishuo Wang
- Department of Obstetrics and Gynecology, Chinese People's Liberation Army Medical School, Beijing, China
| | - Yanmin Ma
- Department of Obstetrics and Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Xiaomeng Bu
- Department of Obstetrics and Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yuexiu Liang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University of Nationalities, Baise, Guangxi, China
| | - Yuzhen Chen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University of Nationalities, Baise, Guangxi, China
| | - Wenpei Bai
- Department of Obstetrics and Gynecology, BeiJing Shijitan Hospital, Beijing, China
| | - Yanrong Chen
- Department of Obstetrics and Gynecology, BeiJing Shijitan Hospital, Beijing, China
| | - Chengyan Deng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| | - Wanyu Zhang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| | - Ming Zhou
- Department of Obstetrics and Gynecology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Lijuan Lv
- Department of Obstetrics and Gynecology, Huanghua Development Zone Boai Hospital, Cangzhou, Hebei, China
| | - Linyan Zhang
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital of Zhuji, Zhuji, Zhejiang, China
| | - Sijia Lu
- Department of Clinical Research, Yikon Genomics Co., Ltd, Suzhou, Jiangsu, China
| | - Wei Shang
- Department of Obstetrics and Gynecology, The Seventh Medical Center of People’s Liberation Army General Hospital, Beijing, China
- Department of Obstetrics and Gynecology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
- Department of Obstetrics and Gynecology, Chinese People's Liberation Army Medical School, Beijing, China
| |
Collapse
|
40
|
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
|
41
|
Huang J, Peng H, Yang D. Research advances in protein lysine 2-hydroxyisobutyrylation: From mechanistic regulation to disease relevance. J Cell Physiol 2024; 239:e31435. [PMID: 39351825 DOI: 10.1002/jcp.31435] [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: 06/06/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 12/18/2024]
Abstract
Histone lysine 2-hydroxyisobutyrylation (Khib) was identified as a novel posttranslational modification in 2014. Significant progress has been made in understanding its roles in reproduction, development, and disease. Although 2-hydroxyisobutyrylation shares some overlapping modification sites and regulatory factors with other lysine residue modifications, its unique structure suggests distinct functions. This review summarizes the latest advancements in Khib, including its regulatory mechanisms, roles in mammalian physiological processes, and its relationship with diseases. This provides direction for further research on Khib and offers new perspectives for developing treatment strategies for related diseases.
Collapse
Affiliation(s)
- Jinglei Huang
- School of Tropical Agriculture and Forestry, Hainan University, Haikou, Hainan, People's Republic of China
| | - Hui Peng
- School of Tropical Agriculture and Forestry, Hainan University, Haikou, Hainan, People's Republic of China
| | - Diqi Yang
- School of Tropical Agriculture and Forestry, Hainan University, Haikou, Hainan, People's Republic of China
| |
Collapse
|
42
|
Wang S, Ren J, Jing Y, Qu J, Liu GH. Perspectives on biomarkers of reproductive aging for fertility and beyond. NATURE AGING 2024; 4:1697-1710. [PMID: 39672897 DOI: 10.1038/s43587-024-00770-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/29/2024] [Indexed: 12/15/2024]
Abstract
Reproductive aging, spanning an age-related functional decline in the female and male reproductive systems, compromises fertility and leads to a range of health complications. In this Perspective, we first introduce a comprehensive framework for biomarkers applicable in clinical settings and discuss the existing repertoire of biomarkers used in practice. These encompass functional, imaging-based and biofluid-based biomarkers, all of which reflect the physiological characteristics of reproductive aging and help to determine the reproductive biological age. Next, we delve into the molecular alterations associated with aging in the reproductive system, highlighting the gap between these changes and their potential as biomarkers. Finally, to enhance the precision and practicality of assessing reproductive aging, we suggest adopting cutting-edge technologies for identifying new biomarkers and conducting thorough validations in population studies before clinical applications. These advancements will foster improved comprehension, prognosis and treatment of subfertility, thereby increasing chances of preserving reproductive health and resilience in populations of advanced age.
Collapse
Affiliation(s)
- Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Jie Ren
- Aging Biomarker Consortium, Beijing, China
- Key Laboratory of RNA Science and Engineering, China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying Jing
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
- Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing Qu
- Aging Biomarker Consortium, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, CAS, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, CAS, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| |
Collapse
|
43
|
Long P, Tan H, Chen B, Wang L, Quan R, Hu Z, Zeng M, Greenbaum J, Shen H, Deng H, Xiao H. Dissecting the shared genetic architecture between anti-Müllerian hormone and age at menopause based on genome-wide association study. Am J Obstet Gynecol 2024; 231:634.e1-634.e11. [PMID: 38969199 PMCID: PMC12038692 DOI: 10.1016/j.ajog.2024.06.050] [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: 10/10/2023] [Revised: 06/10/2024] [Accepted: 06/22/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND While the phenotypic association between anti-Müllerian hormoneand age at menopause has been widely studied, the role of anti-Müllerian hormone in predicting the age at menopause is currently controversial, and the genetic architecture or causal relationships underlying these 2 traits is not well understood. AIM We aimed to explore the shared genetic architecture between anti-Müllerian hormone and age at menopause, to identify shared pleiotropic loci and genes, and to investigate causal association and potential causal mediators. STUDY DESIGN Using summary statistics from publicly available genome-wide association studies on anti-Müllerian hormone (N=7049) and age at menopause (N=201,323) in Europeans, we investigated the global genetic architecture between anti-Müllerian hormone and age at menopause through linkage disequilibrium score regression. We employed pleiotropic analysis under composite null hypothesis, Functional Mapping and Annotation of Genetic Associations, multimarker analysis of GenoMic annotation, and colocalization analysis to identify loci and genes with pleiotropic effects. Tissue enrichment analysis based on Genotype-Tissue Expression data was conducted using the Linkage Disequilibrium Score for the specific expression of genes analysis. Functional genes that were shared were additionally identified through summary data-based Mendelian randomization. The relationship between anti-Müllerian hormone and age at menopause was examined through 2-sample Mendelian randomization, and potential mediators were further explored using colocalization and metabolite-mediated analysis. RESULTS A positive genetic association (correlation coefficient=0.88, P=1.33×10-5) was observed between anti-Müllerian hormone and age at menopause. By using pleiotropic analysis under composite null hypothesis and Functional Mapping and Annotation of Genetic Associations, 42 significant pleiotropic loci were identified that were associated with anti-Müllerian hormone and age at menopause, and 10 of these (rs10734411, rs61913600, rs2277339, rs75770066, rs28416520, rs9796, rs11668344, rs403727, rs6011452, and rs62237617) had colocalized loci. Additionally, 245 significant pleiotropic genes were identified by multimarker analysis of GenoMic annotation. Genetic associations between anti-Müllerian hormone and age at menopause were markedly concentrated in various tissues including whole blood, brain, heart, liver, muscle, pancreas, and kidneys. Further, summary data-based Mendelian randomization analysis revealed 9 genes that may have a causative effect on both anti-Müllerian hormone and age at menopause. A potential causal effect of age at menopause on anti-Müllerian hormone was suggested by 2-sample Mendelian randomization analysis, with very-low-density lipoprotein identified as a potential mediator. CONCLUSION Our study revealed a shared genetic architecture between anti-Müllerian hormone and age at menopause, providing a basis for experimental investigations and individual therapies to enhance reproductive outcomes. Furthermore, our findings emphasized that relying solely on anti-Müllerian hormone is not sufficient for accurately predicting the age at menopause, and a combination of other factors needs to be considered. Exploring new therapeutics aimed at delaying at the onset of menopause holds promise, particularly when targeting shared genes based on their shared genetic architecture.
Collapse
Affiliation(s)
- Panpan Long
- Institute of Reproductive & Stem Cell Engineering, Center of Reproductive Health, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hangjing Tan
- Institute of Reproductive & Stem Cell Engineering, Center of Reproductive Health, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Binbin Chen
- Center of Genetics, Changsha Jiangwan Maternity Hospital, Changsha City, Hunan, China
| | - Le Wang
- Institute of Reproductive & Stem Cell Engineering, Center of Reproductive Health, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Biomedical Research Center, Hunan University of Medicine, Huaihua City, Hunan, China
| | - Ruping Quan
- Institute of Reproductive & Stem Cell Engineering, Center of Reproductive Health, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Zihao Hu
- Institute of Reproductive & Stem Cell Engineering, Center of Reproductive Health, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Minghua Zeng
- Center of Genetics, Changsha Jiangwan Maternity Hospital, Changsha City, Hunan, China
| | - Jonathan Greenbaum
- Deming Department of Medicine, Center of Biomedical Informatics and Genomics, Tulane University School of Medicine, New Orleans, LA
| | - Hui Shen
- Deming Department of Medicine, Center of Biomedical Informatics and Genomics, Tulane University School of Medicine, New Orleans, LA
| | - Hongwen Deng
- Deming Department of Medicine, Center of Biomedical Informatics and Genomics, Tulane University School of Medicine, New Orleans, LA
| | - Hongmei Xiao
- Institute of Reproductive & Stem Cell Engineering, Center of Reproductive Health, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
| |
Collapse
|
44
|
Winstanley YE, Stables JS, Gonzalez MB, Umehara T, Norman RJ, Robker RL. Emerging therapeutic strategies to mitigate female and male reproductive aging. NATURE AGING 2024; 4:1682-1696. [PMID: 39672895 DOI: 10.1038/s43587-024-00771-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/29/2024] [Indexed: 12/15/2024]
Abstract
People today are choosing to have children later in life, often in their thirties and forties, when their fertility is in decline. We sought to identify and compile effective methods for improving either male or female fertility in this context of advanced reproductive age. We found few clinical studies with strong evidence for therapeutics that mitigate reproductive aging or extend fertility; however, this Perspective summarizes the range of emerging experimental strategies under development. Preclinical studies, in mouse models of aging, have identified pharmaceutical candidates that improve egg and sperm quality. Further, a diverse array of medically assisted reproduction methodologies, including those that stimulate rare ovarian follicles and rejuvenate egg quality using mitochondria, may have future utility for older patients. Finally, we highlight the many knowledge gaps and possible future directions in the field of therapeutics to extend the age of healthy human reproduction.
Collapse
Affiliation(s)
- Yasmyn E Winstanley
- Robinson Research Institute, School of Biomedicine; The University of Adelaide, Adelaide, South Australia, Australia
| | - Jennifer S Stables
- Robinson Research Institute, School of Biomedicine; The University of Adelaide, Adelaide, South Australia, Australia
| | - Macarena B Gonzalez
- Robinson Research Institute, School of Biomedicine; The University of Adelaide, Adelaide, South Australia, Australia
| | - Takashi Umehara
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Robert J Norman
- Robinson Research Institute, Adelaide Medical School; The University of Adelaide, Adelaide, South Australia, Australia
| | - Rebecca L Robker
- Robinson Research Institute, School of Biomedicine; The University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
45
|
Benonisdottir S, Straub VJ, Kong A, Mills MC. Genetics of female and male reproductive traits and their relationship with health, longevity and consequences for offspring. NATURE AGING 2024; 4:1745-1759. [PMID: 39672892 DOI: 10.1038/s43587-024-00733-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/26/2024] [Indexed: 12/15/2024]
Abstract
Substantial shifts in reproductive behaviors have recently taken place in many high-income countries including earlier age at menarche, advanced age at childbearing, rising childlessness and a lower number of children. As reproduction shifts to later ages, genetic factors may become increasingly important. Although monogenic genetic effects are known, the genetics underlying human reproductive traits are complex, with both causal effects and statistical bias often confounded by socioeconomic factors. Here, we review genome-wide association studies (GWASs) of 44 reproductive traits of both female and male individuals from 2007 to early 2024, examining reproductive behavior, reproductive lifespan and aging, infertility and hormonal concentration. Using the GWAS Catalog as a basis, from 159 relevant studies, we isolate 37 genes that harbor association signals for four or more reproductive traits, more than half of which are linked to rare Mendelian disorders, including ten genes linked to reproductive-related disorders: FSHB, MCM8, DNAH2, WNT4, ESR1, IGSF1, THRB, BRWD1, CYP19A1 and PTPRF. We also review the relationship of reproductive genetics to related health and behavioral traits, aging and longevity and the effect of parental age on offspring outcomes as well as reflecting on limitations, open questions and challenges in this fast-moving field.
Collapse
Affiliation(s)
- Stefania Benonisdottir
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
- Institute of Physical Science, University of Iceland, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Vincent J Straub
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
| | - Augustine Kong
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
| | - Melinda C Mills
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK.
- Department of Genetics, University Medical Centre Groningen, Groningen, the Netherlands.
- Department of Economics, Econometrics and Finance, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
46
|
Gao H, Ying Y, Sun J, Huang Y, Li X, Zhang D. Genetically Determined Plasma Docosahexaenoic Acid Showed a Causal Association with Female Reproductive Longevity-Related Phenotype: A Mendelian Randomization Study. Nutrients 2024; 16:4103. [PMID: 39683497 DOI: 10.3390/nu16234103] [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: 10/18/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Female reproductive aging remains irreversible. More evidence is needed on how polyunsaturated fatty acids (PUFAs) affect the female reproductive lifespan. OBJECTIVES To identify and validate specific PUFAs that can influence the timing of menarche and menopause in women. METHODS We utilized a two-sample Mendelian randomization (MR) framework to evaluate the causal relationships between various PUFAs and female reproductive longevity, defined by age at menarche (AAM) and age at natural menopause (ANM). Our analyses leveraged summary statistics from four genome-wide association studies (GWASs) on the plasma concentrations of 10 plasma PUFAs, including 8866 to 121,633 European individuals and 1361 East Asian individuals. Large-scale GWASs for reproductive traits provided the genetic data of AAM and ANM from over 202,323 European females and 43,861 East Asian females. Causal effects were estimated by beta coefficients, representing, for each increase in the standard deviation (SD) of plasma PUFA concentration, the yearly increase in AAM or ANM. Replications, meta-analyses, and cross-ancestry effects were assessed to validate the inference. CONCLUSIONS Higher plasma DHA was identified to be associated with delayed natural menopause without affecting menarche, offering a potential intervention target for extending reproductive longevity.
Collapse
Affiliation(s)
- Huajing Gao
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yuewen Ying
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Jing Sun
- Department of Big Data in Health Science, School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yun Huang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xue Li
- Department of Big Data in Health Science, School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang Provincial Birth Defect Control and Prevention Research Center, Hangzhou 310006, China
| |
Collapse
|
47
|
Wang X, Wei Z, Zuo Z, Sun Y, Guo X, Tong Y, Liu G, Xu D, Zhang Z. Sex hormones and reproductive factors with cardiac arrhythmia and ECG indices: a mendelian randomization study. BMC Cardiovasc Disord 2024; 24:659. [PMID: 39567890 PMCID: PMC11577811 DOI: 10.1186/s12872-024-04335-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Sex is a determinant of the incidence and etiology of arrhythmia. Observational and basic studies suggest that sex hormones are essential in this process; however, the relationship between sex hormones and arrhythmia remains unclear. Mendelian randomization (MR) was used to investigate the causal relationships between sex hormone levels, arrhythmia, and electrocardiographic (ECG) indices. METHODS Large genome-wide association studies (GWAS) data on sex hormones, stratified by sex, from the UK biobank consortium, were used as exposure data, and data on atrial fibrillation (AF), atrioventricular block, sick sinus syndrome, paroxysmal tachycardia, and ECG indices were obtained from the FinnGen consortium and summarized large GWAS data. Inverse variance weighting or wald odds was used as the primary analytical method, and weighted medians and MR-Egger regression were used for complementary analyses. The results of the MR of sex hormones and AF from different sources were analyzed using a meta-analysis. Summary-data-based MR analysis was utilized to explore the relationship between sex-hormone related drugs and arrhythmia. RESULTS In men, genetically predicted higher estradiol concentrations were associated with a lower risk of AF (odds ratio: 0.908 [0.852-0.967]; p = 0.0029], whereas genetically predicted higher concentrations of total testosterone were associated with lower heart rate variability. Sex hormones showed no association with atrioventricular block, sick sinus syndrome, paroxysmal tachycardia, resting heart rate, P wave duration, P wave terminal force in lead V1 [PTFV1], PR interval, QRS duration, QTc [QT interval corrected by heart rate], ST duration, spatial [spQRSTa] and frontal [fQRSTa] QRS-T angles in males. In females, there was no significant evidence that sex hormones are associated with arrhythmias or ECG indices. CONCLUSION In this study, we identified a potential causal relationship between estradiol and the risk of AF in males. However, there was no significant association between sex hormones and either arrhythmias or ECG indices in females. These results suggested that sex hormones may play a limited role in cardiac arrhythmias, which requires further verification.
Collapse
Affiliation(s)
- Xishu Wang
- Cardiovascular Medicine Department, Cardiology Center, First Hospital of Jilin University, No.1 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Zhaoyang Wei
- Cardiovascular Medicine Department, Cardiology Center, First Hospital of Jilin University, No.1 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Zheng Zuo
- Cardiovascular Medicine Department, Cardiology Center, First Hospital of Jilin University, No.1 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Ying Sun
- Cardiovascular Medicine Department, Cardiology Center, First Hospital of Jilin University, No.1 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Xiaoxue Guo
- Cardiovascular Medicine Department, Cardiology Center, First Hospital of Jilin University, No.1 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Yi Tong
- Cardiovascular Medicine Department, Cardiology Center, First Hospital of Jilin University, No.1 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Guanghui Liu
- Cardiovascular Medicine Department, Cardiology Center, First Hospital of Jilin University, No.1 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Dongyang Xu
- Cardiovascular Medicine Department, Cardiology Center, First Hospital of Jilin University, No.1 Xinmin Street, Changchun City, 130021, Jilin Province, China
| | - Zhiguo Zhang
- Cardiovascular Medicine Department, Cardiology Center, First Hospital of Jilin University, No.1 Xinmin Street, Changchun City, 130021, Jilin Province, China.
| |
Collapse
|
48
|
Sharma N, Coticchio G, Borini A, Tachibana K, Nasmyth KA, Schuh M. Changes in DNA repair compartments and cohesin loss promote DNA damage accumulation in aged oocytes. Curr Biol 2024; 34:5131-5148.e6. [PMID: 39437784 DOI: 10.1016/j.cub.2024.09.040] [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: 03/08/2024] [Revised: 07/20/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024]
Abstract
Oocyte loss, a natural process that accelerates as women approach their mid-30s, poses a significant challenge to female reproduction. Recent studies have identified DNA damage as a primary contributor to oocyte loss, but the mechanisms underlying DNA damage accumulation remain unclear. Here, we show that aged oocytes have a lower DNA repair capacity and reduced mobility of DNA damage sites compared to young oocytes. Incomplete DNA repair in aged oocytes results in defective chromosome integrity and partitioning, thereby compromising oocyte quality. We found that DNA repair proteins are arranged in spatially distinct DNA repair compartments that form during the late stages of oocyte growth, accompanied by changes in the activity of DNA repair pathways. We demonstrate alterations in these compartments with age, including substantial changes in the levels of key DNA repair proteins and a shift toward error-prone DNA repair pathways. In addition, we show that reduced cohesin levels make aged oocytes more vulnerable to persistent DNA damage and cause changes in DNA repair compartments. Our study links DNA damage accumulation in aged oocytes, a leading cause of oocyte loss, to cohesin deterioration and changes in the organization, abundance, and response of DNA repair machinery.
Collapse
Affiliation(s)
- Ninadini Sharma
- Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | | | - Andrea Borini
- IVIRMA Global Research Alliance, 9.baby, Bologna 40125, Italy
| | - Kikuë Tachibana
- Department of Totipotency, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Munich 82152, Germany
| | - Kim A Nasmyth
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Melina Schuh
- Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany.
| |
Collapse
|
49
|
Biswas L, Tyc KM, Aboelenain M, Sun S, Dundović I, Vukušić K, Liu J, Guo V, Xu M, Scott RT, Tao X, Tolić IM, Xing J, Schindler K. Maternal genetic variants in kinesin motor domains prematurely increase egg aneuploidy. Proc Natl Acad Sci U S A 2024; 121:e2414963121. [PMID: 39475646 PMCID: PMC11551467 DOI: 10.1073/pnas.2414963121] [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: 07/26/2024] [Accepted: 09/27/2024] [Indexed: 11/06/2024] Open
Abstract
The female reproductive lifespan is highly dependent on egg quality, especially the presence of a normal number of chromosomes in an egg, known as euploidy. Mistakes in meiosis leading to egg aneuploidy are frequent in humans. Yet, knowledge of the precise genetic landscape that causes egg aneuploidy in women is limited, as phenotypic data on the frequency of human egg aneuploidy are difficult to obtain and therefore absent in public genetic datasets. Here, we identify genetic determinants of reproductive aging via egg aneuploidy in women using a biobank of individual maternal exomes linked with maternal age and embryonic aneuploidy data. Using the exome data, we identified 404 genes bearing variants enriched in individuals with pathologically elevated egg aneuploidy rates. Analysis of the gene ontology and protein-protein interaction network implicated genes encoding the kinesin protein family in egg aneuploidy. We interrogate the causal relationship of the human variants within candidate kinesin genes via experimental perturbations and demonstrate that motor domain variants increase aneuploidy in mouse oocytes. Finally, using a knock-in mouse model, we validate that a specific variant in kinesin KIF18A accelerates reproductive aging and diminishes fertility. These findings reveal additional functional mechanisms of reproductive aging and shed light on how genetic variation underlies individual heterogeneity in the female reproductive lifespan, which might be leveraged to predict reproductive longevity. Together, these results lay the groundwork for the noninvasive biomarkers for egg quality, a first step toward personalized fertility medicine.
Collapse
Affiliation(s)
- Leelabati Biswas
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Katarzyna M. Tyc
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Mansour Aboelenain
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Department of Theriogenology, Faculty of Veterinary Medicine, Mansoura University, Mansoura35516, Egypt
| | - Siqi Sun
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Iva Dundović
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb1000, Croatia
| | - Kruno Vukušić
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb1000, Croatia
| | - Jason Liu
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | | | - Min Xu
- Department of Statistics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | | | - Xin Tao
- Juno Genetics US, Basking Ridge, NJ07920
| | - Iva M. Tolić
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb1000, Croatia
| | - Jinchuan Xing
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| |
Collapse
|
50
|
Korda M. Menopause age and cancer risk is influenced by rare genetic variants. Nat Struct Mol Biol 2024; 31:1646-1647. [PMID: 39537734 DOI: 10.1038/s41594-024-01434-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
|