1
|
Chen Y, Wang S, Zhang C. The Differentiation Fate of Granulosa Cells and the Regulatory Mechanism in Ovary. Reprod Sci 2025; 32:1414-1426. [PMID: 39192066 DOI: 10.1007/s43032-024-01682-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
Granulosa cells (GCs) are important drives of the reproductive process, not only the supporting cells for nutrition, but also cells with endocrine functions. Their differentiation and development parallel the entire menstruation period and even during pregnancy, making it tightly linked to the fate of the follicle. To elucidate the underlying mechanism is of great significance for related researches. The life course of GCs is briefly divided into five stages, from epithelial cells to pre-granulosa cells, GCs, mural and cumulus cells, lutein cells, and eventually disappear. A wide variety of genes and transcription factors participate in the regulation of different stages, and more importantly, various hormones secreted by the pituitary gland and GCs themselves play a leading role. These endogenous and exogenous signalling molecules interact to form a cross-linked communication network, promoting the development of GCs. Together with oocytes, theca cells and other functional cells in the ovary, GCs drive one of the most vital biological processes in women.
Collapse
Affiliation(s)
- Yilin Chen
- Queen Mary School, Nanchang University, Nanchang, 330006, China
| | - Shimeng Wang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chunping Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
2
|
Li X, Chen X, Guo H. Plasminogen activator inhibitor 1 is a novel predictor in human serum/follicular fluid for diminished ovarian reserve. BMC Womens Health 2025; 25:210. [PMID: 40301911 PMCID: PMC12039065 DOI: 10.1186/s12905-025-03710-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/02/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Diminished ovarian reserve (DOR) is a common female reproductive aging disease, which showed significant impacts on the quality of life and fertility in women. Plasminogen activator inhibitor 1 (PAI-1) is considered to be a major profibrotic factor. The development of DOR is closely related to ovarian fibrosis. The aim of the study was to investigate the expression of PAI-1,which is clinically relevant to DOR. METHODS This case-control study included 40 infertile women with DOR and 40 infertile women with normal ovarian function. PAI-1 and reproductive hormones in serum and follicle fluid were determined in all subjects. Receiver operating characteristic curve (ROC) was applied to evaluate PAI-1 in prediction and diagnosis of DOR. The mRNA and protein expression of PAI-1 in KGN cells induced by cyclophosphamide (CTX) were observed by Western blot (WB) and quantitative real-time PCR (qRT-PCR). RESULTS The sensitivity and specificity of PAI-1 levels in serum/follicular fluid for predicting DOR were 90%/97.5% and 70%/82.5%, respectively. The AUC of PAI-1 in follicular fluid was 0.955(95% CI 0.913-0.997), which cutoff level and Youden index were 68.58 ng/mL and 0.825 for DOR. PAI-1 in serum and follicle fluid showed negative association with Anti-Müllerian hormone (AMH) and antral follicle count (AFC) (serum r= -0.391,r= -0.453;follicle fluid r= -0.486,r= -0.534;p < 0.01), however, they were positively correlated with follicle-stimulating hormone (FSH) and follicle-stimulating hormone/luteinizing hormone (FSH/LH) (serum r = 0.307,r = 0.388;follicle fluid r = 0.300,r = 0.384;p < 0.05). The ROC analysis indicated that serum PAI-1 has great prediction of DOR, with an AUC of 0.841, a sensitivity of 90%, and a specificity of 70%. Additionally, the qRT-PCR results demonstrated that the mRNA levels of PAI-1 increased in the CTX-induced cells (P < 0.05). The western blot results were consistent with qRT-PCR results. CONCLUSION Our study reveals that the expression of PAI-1 is higher in serum and follicular fluid of DOR patients. And it is positively correlated with FSH, FSH/LH and negatively correlated with AMH/AFC, which is necessary to investigate the role of PAI-1 in regulating the growth and development of follicles and the pathogenesis of DOR in future.
Collapse
Affiliation(s)
- Xinshu Li
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xue Chen
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Hua Guo
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
3
|
Wang H. The role of granulosa cells in oocyte development and aging: Mechanisms and therapeutic opportunities. Semin Cell Dev Biol 2025; 171:103614. [PMID: 40300217 DOI: 10.1016/j.semcdb.2025.103614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/20/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025]
Abstract
Granulosa cells (GCs) are essential for oocyte maturation, providing metabolic support, hormonal signaling, and structural integrity critical to successful follicular development. However, advancing age disrupts these functions, driven by factors such as increased oxidative stress, mitochondrial dysfunction, and transcriptomic and proteomic alterations. These age-related changes in GCs contribute to compromised oocyte quality, diminished follicular support, and a decline in fertility, particularly in women of advanced maternal age. This review highlights recent progress in understanding the pivotal roles of GCs in maintaining oocyte health, with a focus on the mechanisms underlying their aging-related dysfunction. Furthermore, we explore promising therapeutic strategies, including antioxidant therapies, metabolic modulators, and GC-based rejuvenation techniques, aimed at mitigating the impacts of reproductive aging. By consolidating and analyzing existing research, this review provides valuable perspectives on fertility preservation and factors shaping reproductive outcomes in women of advanced maternal age.
Collapse
Affiliation(s)
- HaiYang Wang
- Institute of Modern Biology, Nanjing University, Nanjing 210008, China; Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 117411, Singapore.
| |
Collapse
|
4
|
O'Brien CJO. Macrophage Regulation of Hypothalamic-Pituitary-Adrenal and Gonadal Axis Homeostasis and Hormonal Output. Biomed J 2025:100866. [PMID: 40300671 DOI: 10.1016/j.bj.2025.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/17/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025] Open
Abstract
Macrophages are critical immune cells present in virtually every tissue, where they contribute to tissue homeostasis beyond their traditional immune roles. Past and recent evidence highlights their involvement in endocrine regulation, particularly within the hypothalamic-pituitary-adrenal (HPA) and hypothalamic-pituitary-gonadal (HPG) axes. This review explores the ontogeny and function of macrophages residing in the hypothalamus, pituitary, adrenals, and gonads, emphasizing their contributions to hormonal output and endocrine homeostasis. Macrophages in the hypothalamus and pituitary modulate neuroendocrine signalling, impacting stress and reproductive hormone production. In the adrenal glands, distinct macrophage subsets regulate glucocorticoid and mineralocorticoid synthesis, influencing systemic metabolism and blood pressure. Gonadal macrophages contribute to steroidogenesis and fertility, with roles in testosterone production, ovarian folliculogenesis, and corpus luteum maintenance. The emerging understanding of macrophage regulation of endocrine function may seed novel therapeutic approaches for endocrine disorders. Future research should further elucidate the molecular mechanisms underlying macrophage regulation of hormone production and explore their implications for metabolic, immune, and reproductive health.
Collapse
Affiliation(s)
- Conan J O O'Brien
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, 4102, Australia
| |
Collapse
|
5
|
Ge L, Yang Y, Xiao T, Gao Y, Chang W, Du F, Yu M, Zhang JV. Ovarian Endometriosis Accelerates Premature Ovarian Failure and Contributes to Osteoporosis and Cognitive Decline in Aging Mice. Int J Mol Sci 2025; 26:3313. [PMID: 40244208 PMCID: PMC11989598 DOI: 10.3390/ijms26073313] [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: 01/22/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Ovarian endometriosis (OEM) is a chronic inflammatory condition that impairs ovarian function. While its effects on ovarian reserve are well established, the long-term consequences of OEM on ovarian dysfunction, premature ovarian failure (POF), and systemic health, particularly in the context of accelerated aging, remain insufficiently studied. In this study, we employed an OEM mouse model and bulk RNA sequencing to investigate the underlying mechanisms. Our results show that OEM accelerates primordial follicle depletion and upregulates mTOR signaling pathway gene expression, along with mechanical stress and paracrine signaling via the Hippo and Myc pathways. OEM also induces irregular estrus and ovarian fibrosis in aging mice, decreases serum AMH levels, and increases FSH levels. Systemically, elevated serum IgG levels contribute to osteoporosis and cognitive decline, both linked to ovarian dysfunction and POF in OEM. These findings elucidate the mechanisms driving premature ovarian reserve depletion in OEM and highlight its broader systemic effects. This study emphasizes the importance of monitoring ovarian health and ectopic tissue to safeguard ovarian reserves and mitigate long-term risks such as osteoporosis and cognitive decline.
Collapse
Affiliation(s)
- Lei Ge
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
| | - Yali Yang
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
| | - Tianxia Xiao
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
| | - Yuqing Gao
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Wakam Chang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Feifei Du
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
| | - Ming Yu
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
| | - Jian V. Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (L.G.); (M.Y.)
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen 518000, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen 518028, China
- Sino-European Center of Biomedicine and Health, Shenzhen 518000, China
| |
Collapse
|
6
|
Ameho S, Klutstein M. The effect of chronic inflammation on female fertility. Reproduction 2025; 169:e240197. [PMID: 39932461 PMCID: PMC11896653 DOI: 10.1530/rep-24-0197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 02/13/2025]
Abstract
In brief Chronic inflammation causes serious medical conditions in many organs and tissues, including female fertility. Here we review the current literature, showing that chronic inflammation has a negative impact on oocyte quality, folliculogenesis, hormone production, immune signaling and other processes that affect fertility in females. Abstract Inflammation has key biological roles in the battle against pathogens and additional key processes in development and tissue homeostasis. However, when inflammation becomes chronic, it can become a serious medical concern. Chronic inflammation has been shown to contribute to the etiology and symptoms of serious medical conditions such as ulcerative colitis, cardiovascular diseases, endometriosis and various cancers. One of the less recognized symptoms associated with chronic inflammation is its effect on reproduction, specifically on female fertility. Here we review the current literature, showing that chronic inflammation has a negative impact on oocyte quality, folliculogenesis, hormone production, immune signaling and other processes that affect fertility in females. We discuss several factors involved in the etiology of chronic inflammation and its effect on female fertility. We also discuss possible mechanisms by which these effects may be mediated and how interventions may mitigate the effect of chronic inflammation. Finally, we discuss the notion that in many cases, the effect of chronic inflammation is tightly correlated with and resembles the effect of aging, drawing interesting parallels between these processes, possibly through the effect of aging-associated inflammaging.
Collapse
Affiliation(s)
| | - Michael Klutstein
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
7
|
Anderson RA, Marston AL, Telfer EE. Oocyte development: it's all about quality. Reprod Biomed Online 2025; 50:104804. [PMID: 40287201 DOI: 10.1016/j.rbmo.2025.104804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 04/29/2025]
Abstract
Mammalian fertility depends on the production of an oocyte capable of fertilization and supporting early embryo development. This requires both cytoplasmic and nuclear, i.e. chromosomal, competence, processes that were initiated decades prior to ovulation. Current demographic changes with delayed motherhood are increasingly in conflict with these biological processes. This brief review highlights the key stages in oocyte development, as well as recent findings that continue to inform on how the oocyte is able to maintain function over such a prolonged period. These include minimizing oocyte damage caused by the production of reactive oxygen species, the importance of intercellular communication with the surrounding somatic cells, and the molecular mechanisms that underpin the fidelity of chromosome cohesion and then separation at the resumption of meiosis. Some of these are already approaching clinical testing and interventions, with new approaches in the coming years potentially being able to 'put back the clock' to improve oocyte quality.
Collapse
Affiliation(s)
- Richard A Anderson
- Centre for Reproductive Health, Institute for Repair and Regeneration, University of Edinburgh, Edinburgh, UK.
| | - Adele L Marston
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Evelyn E Telfer
- Centre for Reproductive Health, Institute for Repair and Regeneration, University of Edinburgh, Edinburgh, UK; Institute of Cell Biology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
8
|
Wang G, Yang R, Zhang H. Ovarian vascular aging: a hidden driver of mid-age female fertility decline. NPJ AGING 2025; 11:24. [PMID: 40159507 PMCID: PMC11955532 DOI: 10.1038/s41514-025-00216-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Affiliation(s)
- Ge Wang
- College of Biological Sciences, China Agricultural University, 100193, Beijing, China
| | - Ruobing Yang
- College of Biological Sciences, China Agricultural University, 100193, Beijing, China
| | - Hua Zhang
- College of Biological Sciences, China Agricultural University, 100193, Beijing, China.
| |
Collapse
|
9
|
Sun X, Hu X. Unveiling Matrix Metalloproteinase 13's Dynamic Role in Breast Cancer: A Link to Physical Changes and Prognostic Modulation. Int J Mol Sci 2025; 26:3083. [PMID: 40243781 PMCID: PMC11988641 DOI: 10.3390/ijms26073083] [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: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
The biomechanical properties of the extracellular matrix (ECM) including its stiffness, viscoelasticity, collagen architecture, and temperature constitute critical biomechanical cues governing breast cancer progression. Matrix metalloproteinase 13 (MMP13) is an important marker of breast cancer and plays important roles in matrix remodelling and cell metastasis. Emerging evidence highlights MMP13 as a dynamic modulator of the ECM's physical characteristics through dual mechanoregulatory mechanisms. While MMP13-mediated collagen degradation facilitates microenvironmental softening, thus promoting tumour cell invasion, paradoxically, its crosstalk with cancer-associated fibroblasts (CAFs) and tumour-associated macrophages (TAMs) drives pathological stromal stiffening via aberrant matrix deposition and crosslinking. This biomechanical duality is amplified through feedforward loops with an epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) populations, mediated by signalling axes such as TGF-β/Runx2. Intriguingly, MMP13 exhibits context-dependent mechanomodulatory effects, demonstrating anti-fibrotic activity and inhibiting the metastasis of breast cancer. At the same time, angiogenesis and increased metabolism are important mechanisms through which MMP13 promotes a temperature increase in breast cancer. Targeting the spatiotemporal regulation of MMP13's mechanobiological functions may offer novel therapeutic strategies for disrupting the tumour-stroma vicious cycle.
Collapse
Affiliation(s)
- Xiaomeng Sun
- Queen Mary School, Jiangxi Medical College, Nanchang University, Xuefu Avenue, Honggutan District, Nanchang 330031, China;
| | - Xiaojuan Hu
- Queen Mary School, Jiangxi Medical College, Nanchang University, Xuefu Avenue, Honggutan District, Nanchang 330031, China;
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Xuefu Avenue, Honggutan District, Nanchang 330031, China
| |
Collapse
|
10
|
Del Valle JS, Van Helden RW, Moustakas I, Wei F, Asseler JD, Metzemaekers J, Pilgram GSK, Mummery CL, van der Westerlaken LAJ, van Mello NM, Chuva de Sousa Lopes SM. Ex vivo removal of pro-fibrotic collagen and rescue of metabolic function in human ovarian fibrosis. iScience 2025; 28:112020. [PMID: 40104066 PMCID: PMC11914289 DOI: 10.1016/j.isci.2025.112020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/21/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Tissue fibrosis, with the excessive accumulation of extracellular matrix, leads to organ dysfunction. The ovary shows signs of fibrosis from an early age, creating a permissive environment for ovarian cancer. A robust culture-platform to study human ovarian fibrosis would enable screens for antifibrotic drugs to prevent or even reverse this process. Based on previous results showing that androgen therapy can induce ovarian fibrosis, we characterized the fibrotic state of ovaries from transmasculine donors of reproductive age. Anti-inflammatory and antioxidant drugs, such as Pirfenidone, Metformin, and Mitoquinone, could reduce and revert the excess collagen content of the ovarian cortical tissue during culture. We demonstrated that Metformin exerts an antioxidant role and prevents a glycolytic metabolic shift in non-immune ovarian stromal cells in the human ovary, while promoting early folliculogenesis during culture. These results may contribute to develop strategies to manage pro-tumorigenic fibrotic ovarian stroma in advanced age and metabolic disorders.
Collapse
Affiliation(s)
- Julieta S Del Valle
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
| | - Ruben W Van Helden
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
| | - Ioannis Moustakas
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
| | - Fu Wei
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
| | - Joyce D Asseler
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
- Amsterdam UMC, Centre of Expertise on Gender Dysphoria, Amsterdam 1081 HV, the Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam 1081 HV, the Netherlands
| | - Jeroen Metzemaekers
- Department of Gynecology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Gonneke S K Pilgram
- Department of Gynecology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
| | | | - Norah M van Mello
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
- Amsterdam UMC, Centre of Expertise on Gender Dysphoria, Amsterdam 1081 HV, the Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam 1081 HV, the Netherlands
| | - Susana M Chuva de Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
- Ghent-Fertility and Stem Cell Team (G-FAST), Department of Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
11
|
Winstanley YE, Rose RD, Sobinoff AP, Wu LL, Adhikari D, Zhang QH, Wells JK, Wong LH, Szeto HH, Piltz SG, Thomas PQ, Febbraio MA, Carroll J, Pickett HA, Russell DL, Robker RL. Telomere length in offspring is determined by mitochondrial-nuclear communication at fertilization. Nat Commun 2025; 16:2527. [PMID: 40087268 PMCID: PMC11909127 DOI: 10.1038/s41467-025-57794-7] [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: 06/15/2023] [Accepted: 03/02/2025] [Indexed: 03/17/2025] Open
Abstract
The initial setting of telomere length during early life in each individual has a major influence on lifetime risk of aging-associated diseases; however there is limited knowledge of biological signals that regulate inheritance of telomere length, and whether it is modifiable is not known. We now show that when mitochondrial activity is disrupted in mouse zygotes, via exposure to 20% O2 or rotenone, telomere elongation between the 8-cell and blastocyst stage is impaired, with shorter telomeres apparent in the pluripotent Inner Cell Mass (ICM) and persisting after organogenesis. Identical defects of elevated mtROS in zygotes followed by impaired telomere elongation, occurred with maternal obesity or advanced age. We further demonstrate that telomere elongation during ICM formation is controlled by mitochondrial-nuclear communication at fertilization. Using mitochondrially-targeted therapeutics (BGP-15, MitoQ, SS-31, metformin) we demonstrate that it is possible to modulate the preimplantation telomere resetting process and restore deficiencies in neonatal telomere length.
Collapse
Affiliation(s)
- Yasmyn E Winstanley
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Ryan D Rose
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- Genea Fertility SA, St. Andrews Hospital, Adelaide, SA, Australia
| | - Alexander P Sobinoff
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Linda L Wu
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Deepak Adhikari
- Development and Stem Cells Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Qing-Hua Zhang
- Development and Stem Cells Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Jadon K Wells
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Lee H Wong
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne VIC, Australia
| | | | - Sandra G Piltz
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Paul Q Thomas
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - John Carroll
- Development and Stem Cells Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Hilda A Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Darryl L Russell
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Rebecca L Robker
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia.
- Development and Stem Cells Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
12
|
Lee DR, Lee JE. Preservation of ovarian function using human pluripotent stem cell-derived mesenchymal progenitor cells. Clin Exp Reprod Med 2025; 52:30-37. [PMID: 38782038 PMCID: PMC11900672 DOI: 10.5653/cerm.2024.07003] [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: 02/24/2024] [Revised: 04/03/2024] [Accepted: 04/06/2024] [Indexed: 05/25/2024] Open
Abstract
Ovarian reserve diminishes with age, and older women experience a corresponding shift in sex hormone levels. These changes contribute to an age-dependent decrease in fertility and a decline in overall health. Furthermore, while survival rates following cancer treatment have improved for young female patients, a reduction in ovarian function due to the side effects of such treatments can be difficult to avoid. To date, no effective therapy has been recommended to preserve ovarian health in these patients. Mesenchymal progenitor cells (MPCs) are considered a promising option for cell therapy aimed at maintaining fertility and fecundity. Although MPCs derived from human adult tissues are recognized for their various protective effects against ovarian senescence, they are limited in quantity. Consequently, human pluripotent stem cell-derived MPCs (hPSC-MPCs), which exhibit high proliferative capacity and retain genetic stability during growth, have been utilized to delay reproductive aging. This review highlights the impact of hPSC-MPCs on preserving the functionality of damaged ovaries in female mouse models subjected to chemotherapy and natural aging. It also proposes their potential as a valuable cell source for fertility preservation in women with a variety of diseases.
Collapse
Affiliation(s)
- Dong Ryul Lee
- Department of Biomedical Science, College of Biological Science, CHA University, Seongnam, Republic of Korea
- Department of Biochemisty, CHA University School of Medicine, Seongnam, Republic of Korea
- CHA Advanced Research Institute, CHA Bundang Medical Center, Seongnam, Republic of Korea
| | - Jeoung Eun Lee
- CHA Advanced Research Institute, CHA Bundang Medical Center, Seongnam, Republic of Korea
| |
Collapse
|
13
|
Fan H, Li C, Lv Y, Qi T, Huang Y, Ma L, Lan Y, Chen P, Lou Z, Zhou J. Tetramethyl bisphenol F exposure induces human ovarian granulosa cell senescence and mouse ovarian damage by regulating ESRRB signalling. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117940. [PMID: 40014986 DOI: 10.1016/j.ecoenv.2025.117940] [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: 12/07/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Tetramethyl bisphenol F (TMBPF), regarded as a promising alternative to bisphenol A (BPA) across multiple industries, is now incorporated into polymer coatings for metal cans used in food and beverage packaging. Recent research has highlighted possible health risks associated with TMBPF, yet its impact on ovarian function and the mechanisms involved remain inadequately understood. This study provides a thorough evaluation of TMBPF's effects on granulosa cells (GCs) and uncovers its transgenerational influence on female reproductive health. We exposed pregnant CD1 mice to TMBPF at doses of 50 μg/kg/day or 200 μg/kg/day from gestational day 7 (GD7) through postnatal day 21 (PND21). Our findings indicated that maternal exposure to TMBPF adversely affects the reproductive system of the F1 generation by elevating the levels of E2, FSH, and LH, reducing the ovarian index, and increasing the number of collagen fibers in the ovarian stroma. The human granulosa-like tumor cell line KGN was employed as an experimental model to assess the toxic effects of TMBPF on GCs. The findings revealed that sublethal concentrations of TMBPF induced mitochondrial dysfunction, cellular senescence, and disrupted redox homeostasis in GCs in vitro. Our transcriptome sequencing analyses indicated that ESRRB upregulation might play a critical role in TMBPF-induced cellular senescence. Additionally, TMBPF exposure induced KGN cells senescence and senescent cell accumulation in F1 mouse ovaries by promoting ESRRB/p21 signalling. Collectively, our results indicate that TMBPF disrupts hormone levels in the ovaries, accelerates GCs senescence and promotes ovarian fibrosis, adversely affecting ovarian function in offspring. This study provides new insights into the potential effects of TMBPF exposure on the reproductive system and the related mechanisms.
Collapse
Affiliation(s)
- Huiyu Fan
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Key Laboratory of Women's Reproductive Health of Zhejiang Province, Hangzhou, PR China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
| | - Chunming Li
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
| | - Yifei Lv
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Key Laboratory of Women's Reproductive Health of Zhejiang Province, Hangzhou, PR China; Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Tongyun Qi
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Key Laboratory of Women's Reproductive Health of Zhejiang Province, Hangzhou, PR China; Department of Gynecology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Yizhou Huang
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
| | - Linjuan Ma
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
| | - Yibing Lan
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
| | - Peiqiong Chen
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
| | - Zheng Lou
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Key Laboratory of Women's Reproductive Health of Zhejiang Province, Hangzhou, PR China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
| | - Jianhong Zhou
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China.
| |
Collapse
|
14
|
Liu Z, Zhou Q, Zan J, Tian J, Zhang Y, Wu F, Zhao H, Peng Q, Liu S, Chen Q, Liu E, Liao Z, Zou P, Mei L, Wang W, Dong S, Niu L, Wu S, He L, Zhou X, Jin Y, Li P, Yang S. Proteomic Analysis of Human Follicular Fluid-Derived Exosomes Reveals That Insufficient Folliculogenesis in Aging Women is Associated With Infertility. Mol Cell Proteomics 2025; 24:100930. [PMID: 40024376 PMCID: PMC11994977 DOI: 10.1016/j.mcpro.2025.100930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 09/28/2024] [Accepted: 12/21/2024] [Indexed: 03/04/2025] Open
Abstract
Although the risk of female infertility increases with advancing age, the underlying mechanisms remain unknown. Exosomes in follicular fluid are suggested to regulate folliculogenesis and influence oocyte quality, potentially playing a critical role in age-related infertility. Elucidating their content could enhance the understanding of the molecular mechanisms associated with female aging-induced infertility. In this study, we explored the proteomic profiles of exosomes derived from human follicular fluid to identify protein signatures associated with infertility in both young and aging women. Despite the lack of significant differences in the morphology and particle size of follicular fluid-derived exosomes between the two groups, proteomic analysis revealed a distinct pattern of differentially expressed proteins (DEPs). DEPs associated with B-cell activation, pathogen invasion, and disrupted metabolic processes were significantly more highly expressed in the aging group than in the young group, indicating their involvement in age-related infertility. In vivo experiments demonstrated that the application of exosomes, particularly those derived from young female group, facilitated the successful maturation of follicles. Key exosomal proteins, including ENO1, HSP90B1, fetuin-B, C7, and APOC4, were found to be associated with follicular maturation. Furthermore, the PI3K/AKT signaling pathway, which is known to be related to folliculogenesis, was activated by the application of exosomes in aging female mice. This study provides novel insights into the aging-associated protein signatures of follicular fluid-derived exosomes and their potential role in infertility. These findings suggest that aging-related protein signatures in exosomes could contribute to the treatment of age-related infertility.
Collapse
Affiliation(s)
- Zhen Liu
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Qilin Zhou
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Jun Zan
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang, China
| | - Jingyan Tian
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Yangzhuohan Zhang
- Department of Clinical Medicine, Hubei University of Science and Technology, Xianning, China
| | - Fanggui Wu
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Huan Zhao
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Qianwen Peng
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Shangjie Liu
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Qianjun Chen
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Endong Liu
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Zhengdong Liao
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Pengfei Zou
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Lin Mei
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Wen Wang
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Sen Dong
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Luo Niu
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Shengda Wu
- Shenzhen University Medical School, Shenzhen University General Hospital, Shenzhen, China
| | - Liangge He
- Shenzhen University Medical School, Shenzhen University General Hospital, Shenzhen, China
| | - Xiaoyi Zhou
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Yanbo Jin
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Panpan Li
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Sheng Yang
- The Reproductive Medicine Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China.
| |
Collapse
|
15
|
Wang H, Yang L. Ovarian Mechanobiology: Understanding the Interplay Between Mechanics and Follicular Development. Cells 2025; 14:355. [PMID: 40072084 PMCID: PMC11898978 DOI: 10.3390/cells14050355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
The ovary is a dynamic organ where mechanical forces profoundly regulate follicular development, oocyte maturation, and overall reproductive function. These forces, originating from the extracellular matrix (ECM), granulosa and theca cells, and ovarian stroma, influence cellular behavior through mechanotransduction, translating mechanical stimuli into biochemical responses. This review explores the intricate interplay between mechanical cues and ovarian biology, focusing on key mechanosensitive pathways such as Hippo signaling, the PI3K/AKT pathway, and cytoskeletal remodeling, which govern follicular dormancy, activation, and growth. Additionally, it examines how ovarian aging disrupts the mechanical microenvironment, with ECM stiffening and altered mechanotransduction contributing to a decline in ovarian reserve and reproductive potential. Emerging technologies, including 3D culture systems and organ-on-chip platforms, are highlighted for their ability to replicate the ovarian microenvironment and advance drug discovery and therapeutic interventions. By integrating mechanobiological principles, this review aims to enhance our understanding of ovarian function and provide new strategies for preserving fertility and combating infertility.
Collapse
Affiliation(s)
- Haiyang Wang
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Liuqing Yang
- NUS Bia-Echo Asia Centre of Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
16
|
Zhang JK, Li XP, Tang Y, Zeng LP, Liu X, Zhang JL, Chen CY, Zheng S, Liu ZZ, Gong X, Jose PA, Guo L, Zeng CY. Semaglutide ameliorates metabolic disorders in offspring via regulation of oocyte ROS of pre-pregnancy obesity mice. Acta Pharmacol Sin 2025:10.1038/s41401-025-01501-1. [PMID: 39984623 DOI: 10.1038/s41401-025-01501-1] [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: 07/11/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025]
Abstract
Pre-pregnancy obesity (PPO) seriously threatens the health of both mother and offspring. Pre-pregnancy weight management is particularly important for the prevention of metabolic diseases in offspring. Semaglutide is one of the most effective glucagon-like peptide-1 agonizts for the management of obesity and metabolic diseases, but little is known about its effect on the long-term health of offspring. In this study we investigated the effects of semaglutide administered before pregnancy on the offspring health from PPO mice. PPO mice model was established by feeding with high-fat diet for 16 weeks, and then injected with semaglutide (30 nmol/kg-1·d-1, sc.) for 22 days before pregnancy. After the treatment, the mice were mated with normal males or underwent in vitro fertilization (IVF) for offspring reproduction. We showed that the semaglutide treatment not only improved the lipid and glucose metabolic disorders and fertility of PPO mice, but also significantly reversed the overweight, impaired energy balance, adipose inflammatory state, lipid and glucose metabolic disorders and insulin resistance of their IVF offspring. By conducting RNA-seq analysis, SOD activity and malondialdehyde assays in ovaries, as well as ROS staining in oocytes, we revealed that the semaglutide treatment reduced the elevated oxidative stress in ovaries and high ROS levels in oocytes from PPO mice, possibly through activating the PI3K/AKT pathway and improving the state of SOD. Interestingly, incubation of oocytes from semaglutide-treated dams with H2O2 (100 μM) in vitro during IVF blocked the protective effects of semaglultide against the metabolic disorders in the offspring. In conclusion, semaglutide treatment before pregnancy effectively alleviates obesity-related metabolic disorders in offspring. The regulation of ROS in oocytes plays a crucial role in the protective effects of semaglutide.
Collapse
Affiliation(s)
- Jun-Kai Zhang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Department of Cardiology, Sichuan Provincial Corps Hospital of Chinese People's Armed Police Force (Sichuan Corps Hospital of PAP), Leshan, 614000, China
| | - Xiao-Ping Li
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Department of Cardiology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410081, China
| | - Yang Tang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Li-Ping Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xuan Liu
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Jian-Li Zhang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Cai-Yu Chen
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Zhi-Zhao Liu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Xue Gong
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, The George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Li Guo
- Endocrinology Department, The First Affiliated Hospital of The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Chun-Yu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China.
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China.
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
17
|
Zhang K, Xu R, Ma J, Zheng L, Zhang H, Li Z, Weng H, Yang X, Hu Y, Chen X, Shu J. Pyrroloquinoline-quinone supplementation restores ovarian function and oocyte quality in a mouse model of advanced maternal age†. Biol Reprod 2025; 112:346-360. [PMID: 39579343 DOI: 10.1093/biolre/ioae174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 11/25/2024] Open
Abstract
Natural ovarian aging is one of the major causes for declining fertility in female animals, which has become an insurmountable issue in human reproduction clinics and assisted reproductive technology procedures. Nevertheless, the molecular basis of oocyte aging remains poorly understood, and feasible improvement strategies are unavailable. In the present study, in vivo supplementation of pyrroloquinoline-quinone effectively elevated the fecundity of reproductively aged mice by balancing hormonal secretion, harmonizing the estrus cycle, and eliminating ovarian fibrosis. Moreover, oocyte quality also increased in aged mice after pyrroloquinoline-quinone administration from various aspects, including nuclear and cytoplasmic maturation competency, fertilization capacity, and pre-implantation embryonic development potential. Transcriptomic analysis identified target pathways that might mediate pyrroloquinoline-quinone's effects in aged oocytes. Specifically, it was demonstrated that pyrroloquinoline-quinone supplementation restored the mitochondrial dynamics and lysosomal function to remove excessive reactive oxygen species and suppress apoptosis in aged oocytes. Jointly, these findings demonstrate pyrroloquinoline-quinone administration is an efficacious method to restore the compromised ovary function and damaged oocyte quality in reproductively aged mice, which might be a potential clinical therapy for women of advanced maternal age with infertility.
Collapse
Affiliation(s)
- Kemei Zhang
- Reproductive Medicine Center, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
- Department of Reproductive Medicine, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Rui Xu
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jinzhao Ma
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lu Zheng
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hong Zhang
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhou Li
- Department of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hong Weng
- School of Pharmaceutical Sciences, Zhejiang Pharmaceutical University, Ningbo, China
| | - Xiaoyue Yang
- School of Pharmaceutical Sciences, Zhejiang Pharmaceutical University, Ningbo, China
| | - Ying Hu
- School of Pharmaceutical Sciences, Zhejiang Pharmaceutical University, Ningbo, China
| | - Xueqing Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Jing Shu
- Reproductive Medicine Center, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| |
Collapse
|
18
|
Gonzalez MB, Rose RD, Connaughton HS, Mackintosh G, Bugeja C, Barry M, McPherson NO, Robker RL. Sperm oxidative damage acquired during seminal plasma removal for assisted reproductive technology is reduced by BGP-15. J Assist Reprod Genet 2025:10.1007/s10815-025-03418-4. [PMID: 39934465 DOI: 10.1007/s10815-025-03418-4] [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: 12/05/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
PURPOSE Semen manipulation for assisted reproductive technology (ART) causes spermatozoa damage; thus, we investigated the potential of the novel therapeutic BGP-15 to preserve sperm quality during semen washing prior to insemination. METHODS Donated human ejaculates (N = 40), with or without 10 µM BGP-15, were analyzed for sperm motility, DNA fragmentation, and oxidation. Seminal plasma was removed using different clinical sperm selection methods: simple wash, swim-up, or density gradient centrifugation (DGC), followed by assessment for sperm motility, mitochondrial ROS (mtROS), mitochondrial membrane potential (MMP), and DNA fragmentation and oxidation. RESULTS Donated semen samples incubated with BGP-15 had increased sperm motility (+ 15%, p = 0.002) and reduced oxidative DNA damage levels (- 57%, p = 0.03). Samples processed by simple wash had the highest sperm count compared with DGC (+ 55%, p < 0.005) and swim-up (+ 21%, p < 0.0005). Swim-up showed increased vitality compared with DGC (+ 18%, p < 0.001) and simple wash (+ 27%, p < 0.0001), as well as the lowest DNA oxidation levels compared with simple wash - 40%, (p = 0.01) and DGC (- 76%, p < 0.0001). Swim-up also had the lowest mitochondrial membrane potential compared with simple wash and DGC (- 28%, p < 0.03). Comparison between untreated and BGP-15-treated groups for each sperm washing method showed that BGP-15 increased MMP in DGC sperm (+ 11%, p = 0.0006), and reduced DNA fragmentation in washed samples (- 22%, p = 0.03). Moreover, BGP-15 lowered DNA oxidation in all preparation methods: washed (- 48%, p = 0.002), swim-up (- 42%, p = 0.04), and DGC (- 29%, p < 0.0001). CONCLUSIONS The inclusion of BGP-15 during semen preparation can protect sperm quality and, in the future, may be used clinically to improve sperm selection methods.
Collapse
Affiliation(s)
- Macarena B Gonzalez
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia.
| | - Ryan D Rose
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia
- Genea Fertility SA, St. Andrews Hospital, South Terrace, Adelaide, SA, 5000, Australia
| | - Haley S Connaughton
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Gracie Mackintosh
- Genea Fertility SA, St. Andrews Hospital, South Terrace, Adelaide, SA, 5000, Australia
| | - Caitlyn Bugeja
- Genea Fertility SA, St. Andrews Hospital, South Terrace, Adelaide, SA, 5000, Australia
| | - Michael Barry
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia
- Genea Fertility SA, St. Andrews Hospital, South Terrace, Adelaide, SA, 5000, Australia
| | - Nicole O McPherson
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia
- Genea Fertility SA, St. Andrews Hospital, South Terrace, Adelaide, SA, 5000, Australia
- Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Rebecca L Robker
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia
| |
Collapse
|
19
|
Pan X, Ren Z, Gong P, Sun X, Wang L, He Y, Liu D, Cao X. Cryptotanshinone Inhibits Obesity-Related Cervical Cancer by Downregulating CXCL8 Expression in Hela Cells. Chem Biodivers 2025:e202403263. [PMID: 39921242 DOI: 10.1002/cbdv.202403263] [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] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
Cervical cancer is one of the cancers commonly found in the female reproductive system and is associated with obesity. However, the exact connection mechanisms remain unclear. Screening of key therapeutic targets and natural products with good antitumor activity has become a crucial strategy for cancer therapy. Cryptotanshinone is one of the main extracts of tanshinone, which has anti-inflammatory and anti-cancer properties. Key therapeutic targets and related low-toxicity natural active ingredients were identified as crucial components in cancer treatment strategies. Therefore, network pharmacology and cellular biology techniques were used to screen and validate key targets in obesity-related cervical cancer and to elucidate the mechanisms of its cryptotanshinone. The results indicated that C-X-C motif chemokine ligand 8 (CXCL8) might be modulated by cryptotanshinone. The knockdown of CXCL8 significantly reduced Hela cell viability to 15.29% ± 4.59% compared with the control group (p < 0.01), which consequently inhibited both cell proliferation and lipid droplet formation. Moreover, cryptotanshinone (20, 40, and 80 µM) significantly reduced CXCL8 expression and inhibited the NOD-like receptor signaling pathway in Hela cells compared with the control group (p < 0.01). Therefore, this study manifested that cryptotanshinone potentially played an important role in obesity-related cervical cancer. This study provided an important experimental basis for further exploring the pathogenesis and prevention of obesity-related cervical cancer.
Collapse
Affiliation(s)
- Xinyu Pan
- Department of Bioinformatics, School of Life Science, Liaoning University, Shenyang, China
| | - Ziqi Ren
- Department of Bioinformatics, School of Life Science, Liaoning University, Shenyang, China
| | - Ping Gong
- Hunan Institute of Microbiology, Changsha, China
| | - Xiaodan Sun
- Department of Bioinformatics, School of Life Science, Liaoning University, Shenyang, China
| | - Le Wang
- Department of Bioinformatics, School of Life Science, Liaoning University, Shenyang, China
| | - Yin He
- Department of Bioinformatics, School of Life Science, Liaoning University, Shenyang, China
| | - Dan Liu
- Department of Bioinformatics, School of Life Science, Liaoning University, Shenyang, China
| | - Xiangyu Cao
- Department of Bioinformatics, School of Life Science, Liaoning University, Shenyang, China
| |
Collapse
|
20
|
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
|
21
|
Gonzalez MB, McPherson NO, Connaughton HS, Winstanley YE, Kennedy DT, Campugan CA, Febbraio MA, Barry M, Rose RD, Robker RL. Mitochondrial activator BGP-15 protects sperm quality against oxidative damage and improves embryo developmental competence. F&S SCIENCE 2025; 6:42-54. [PMID: 39675561 DOI: 10.1016/j.xfss.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE To study the efficacy of mitochondrial activator BGP-15 to preserve sperm quality and competence against cellular damage. DESIGN Spermatozoa from mice or humans were treated in vitro with BGP-15, and sperm quality markers were assessed. Spermatozoa from young (8-12 weeks old) or reproductively old (>14 months old) mice were treated with BGP-15 for 1 hour and assessed for sperm quality and preimplantation embryo development after in vitro fertilization. The safety of BGP-15 on offspring outcomes was assessed through embryo transfers. In parallel studies, spermatozoa from healthy (not infertile) men were incubated in hydrogen peroxide, to induce oxidative stress, plus increasing doses of BGP-15, and sperm quality was evaluated. Spermatozoa from patients undergoing assisted reproductive technology (ART) treatment were incubated in the optimized dose of BGP-15 for 30 minutes, and sperm quality was assessed. SUBJECTS C57BL/6 mice (N = 4-15 per group) for sperm quality and embryo development. CBAF1 mice (n = 6 per group) produced embryos for transfer. Human spermatozoa were from men with no infertility diagnosis (n = 14-20) or men undergoing ART (n = 33) at a local fertility clinic. EXPOSURE Mouse spermatozoa were treated with 10-μM BGP-15. Human spermatozoa were treated with BGP-15 at doses from 1 to 100 μM. MAIN OUTCOME MEASURES Sperm quality measures (mouse and human) included motility, mitochondrial membrane potential (JC-1 dye), deoxyribonucleic acid (DNA) fragmentation ("HALO" assay), and DNA oxidation (8-oxoguanine immunodetection). Mouse embryo and offspring measures included on-time development after in vitro fertilization, morphokinetic analysis, and blastocyst inner cell mass and trophectoderm cell number, and growth and development from birth to 21 days postnatally. RESULTS BGP-15 increased sperm motility and mitochondrial membrane potential and decreased DNA oxidation in old mice. BGP-15 improved on-time development of 2-cell and blastocyst embryos and increased the inner cell mass blastomere number. Embryos from BGP-15-treated mouse spermatozoa produced normal offspring. In human spermatozoa subjected to in vitro oxidative stress, BGP-15 increased motility by 45% and prevented DNA fragmentation (by 45%) and oxidative damage (by 60%). In spermatozoa from men attending a fertility clinic, BGP-15 increased motility by 12% and reduced both DNA oxidation and fragmentation by >20%. CONCLUSION BGP-15 protects sperm against cellular damage and has the potential to improve ART outcomes.
Collapse
Affiliation(s)
- Macarena B Gonzalez
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Nicole O McPherson
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Genea Fertility SA, Adelaide, South Australia, Australia; Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, South Australia, Australia
| | - Haley S Connaughton
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Yasmyn E Winstanley
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - David T Kennedy
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Carl A Campugan
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Michael Barry
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Genea Fertility SA, Adelaide, South Australia, Australia
| | - Ryan D Rose
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Genea Fertility SA, Adelaide, South Australia, Australia
| | - Rebecca L Robker
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
22
|
Zhang K, Xu R, Zheng L, Zhang H, Qian Z, Li C, Xue M, He Z, Ma J, Li Z, Chen L, Ma R, Yao B. Elevated N-glycosylated cathepsin L impairs oocyte function and contributes to oocyte senescence during reproductive aging. Aging Cell 2025; 24:e14397. [PMID: 39494952 PMCID: PMC11822660 DOI: 10.1111/acel.14397] [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: 09/04/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024] Open
Abstract
Age-related declines in oocyte quality and ovarian function are pivotal contributors to female subfertility in clinical settings. Yet, the mechanisms driving ovarian aging and oocyte senescence remain inadequately understood. The present study evaluated the alterations in N-glycoproteins associated with ovarian aging and noted a pronounced elevation in N221 glycopeptides of cathepsin L (Ctsl) in the ovaries of reproductive-aged mice (8-9 months and 11-12 months) compared to younger counterparts (6-8 weeks). Subsequent analysis examined the involvement of Ctsl in oocyte aging and demonstrated a significant elevation in Ctsl levels in aged oocytes. Further, it was revealed that the overexpression of Ctsl in young oocytes substantially diminished their quality, while oocytes expressing an N221-glycosylation mutant of Ctsl did not suffer similar quality degradation. This finding implies that the N221 glycosylation of Ctsl is pivotal in modulating its effect on oocyte health. The introduction of a Ctsl inhibitor into the culture medium restored oocyte quality in aged oocytes by enhancing mitochondrial function, reducing accumulated reactive oxygen species (ROS), lowering apoptosis, and recovering lysosome capacity. Furthermore, the targeted downregulation of Ctsl using siRNA microinjection in aged oocytes enhanced fertilization capability and blastocyst formation, affirming the role of Ctsl knockdown in fostering oocyte quality and embryonic developmental potential. In conclusion, these findings underscore the detrimental effects of high expression of N-glycosylated Ctsl on oocyte quality and its contribution to oocyte senescence, highlighting it as a potential therapeutic target to delay ovarian aging and enhance oocyte viability.
Collapse
Affiliation(s)
- Kemei Zhang
- Department of Reproductive MedicineJinling Clinical Medical College, Nanjing Medical UniversityNanjingChina
| | - Rui Xu
- Department of Reproductive MedicineJinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Lu Zheng
- Department of Reproductive MedicineJinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Hong Zhang
- Department of Reproductive MedicineJinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Zhang Qian
- Department of Reproductive MedicineJinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Chuwei Li
- Department of Reproductive MedicineJinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Mengqi Xue
- Department of Reproductive MedicineJinling Clinical Medical College, Nanjing Medical UniversityNanjingChina
| | - Zhaowanyue He
- Department of Reproductive MedicineJinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Jinzhao Ma
- Department of Reproductive MedicineJinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Zhou Li
- Department of Reproductive MedicineJinling Hospital, School of Medicine, Jiangsu UniversityZhenjiangChina
| | - Li Chen
- Department of Reproductive MedicineJinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Rujun Ma
- Department of Reproductive MedicineJinling Clinical Medical College, Nanjing Medical UniversityNanjingChina
- Department of Reproductive MedicineJinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Bing Yao
- Department of Reproductive MedicineJinling Clinical Medical College, Nanjing Medical UniversityNanjingChina
- Department of Reproductive MedicineJinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
- Department of Reproductive MedicineJinling Hospital, School of Medicine, Jiangsu UniversityZhenjiangChina
| |
Collapse
|
23
|
Zhu Z, Lei M, Guo R, Xu Y, Zhao Y, Wei C, Yang Q, Sun Y. Nicotinamide riboside supplementation ameliorates ovarian dysfunction in a PCOS mouse model. J Ovarian Res 2025; 18:9. [PMID: 39833950 PMCID: PMC11749135 DOI: 10.1186/s13048-025-01596-4] [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: 09/19/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is the leading cause of anovulatory infertility among women of reproductive age, yet the range of effective treatment options remains limited. Our previous study revealed that reduced levels of nicotinamide adenine dinucleotide (NAD+) in ovarian granulosa cells (GCs) of women with PCOS resulted in the accumulation of reactive oxygen species (ROS) and mitochondrial dysfunction. However, it is still uncertain whether increasing NAD+ levels in the ovaries could improve ovarian function in PCOS. In this study, we demonstrated that supplementation with the NAD+ precursor nicotinamide riboside (NR) prevented the decrease in ovarian NAD+ levels, normalized estrous cycle irregularities, and enhanced ovulation potential in dehydroepiandrosterone (DHEA)-induced PCOS mice. Moreover, NR supplementation alleviated ovarian fibrosis and enhanced mitochondrial function in ovarian stromal cells of PCOS mice. Furthermore, NR supplementation improved oocyte quality in PCOS mice, as evidenced by reduced abnormal mitochondrial clustering, enhanced mitochondrial membrane potential, decreased ROS levels, reduced spindle abnormality rates, and increased early embryonic development potential in fertilized oocytes. These findings suggest that supplementing with NAD+ precursors could be a promising therapeutic strategy for addressing ovarian infertility associated with PCOS.
Collapse
Affiliation(s)
- Zhenye Zhu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Min Lei
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruizhi Guo
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yining Xu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanqing Zhao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chenlu Wei
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingling Yang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
24
|
Sarkar P, Moore M, Ozmen A, Cetinkaya-Un B, Julie V, Imudia AN, Lockwood CJ, Kayisli UA, Guzeloglu-Kayisli O. Enhanced ovarian FKBP51 expression is associated with ovarian aging: a molecular insight for age-related fertility in women. F&S SCIENCE 2025:S2666-335X(25)00004-7. [PMID: 39837475 DOI: 10.1016/j.xfss.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE To study the relationship between FK506-binding protein 51 (FKBP51) and ovarian aging and/or diminished ovarian reserve (DOR) in human ovaries by comparing FKBP51 levels in granulosa cells (GCs) and cumulus cells (CCs), collected during controlled ovarian stimulation (COS) from women of advanced reproductive age and/or with a diagnosis of DOR with that of young women with normal ovarian reserve. To explore the association between increased FKBP51 expression and human ovarian aging further, expression of FKBP51 was compared in ovarian stroma of postmenopausal vs. premenopausal women. Lastly, this relation was further queried by comparing ovarian expression of several collagen genes as markers of ovarian fibrosis in 14-month-old wild-type (Fkbp5+/+) and Fkbp5 knockout (Fkbp5-/-) mice. DESIGN Laboratory-based experimental study. SUBJECTS Samples collected included follicular fluid, CCs, GCs, and serum from group 1: young women with normal ovarian reserve (<35 years; n = 12); group 2: DOR (antimüllerian hormone <1 ng/mL; n = 10); and group 3: women of advanced age with normal ovarian reserve (>37 years; n = 8). Ovarian stromal tissues obtained from surgical specimen of post-menopausal (50-65 years; n = 6) and pre-menopausal (18-30 years; n = 6). Ovarian tissues from 14-month-old Fkbp5+/+and Fkbp5-/- mice. All the experiments were performed at an academic-affiliated assisted reproductive technology unit/laboratory. EXPOSURE Comparison of FKBP51 expression in GCs and CCs from women undergoing COS, ovarian stromal tissue from pre- and post-menopausal women, and ovarian tissue from aged Fkbp5+/+and Fkbp5-/- mice. MAIN OUTCOME MEASURES (1) Level of FKBP51 in human GCs and CCs, collected during COS by performing real-time quantitative polymerase chain reaction (qPCR). (2) Immunohistochemistry to detect FKBP51 levels and Picrosirius Red staining to detect collagen deposition in human ovarian stromal tissue. (3) Real-time qPCR to compare expression levels of several collagen genes in Fkbp5+/+ and Fkbp5-/- old mice ovaries. Serum and follicular fluid levels of transforming growth factor β1, and soluble endoglin measured by enzyme-linked immunosorbent assay. RESULTS Immunohistochemistry revealed that FKBP51 histologic score levels in ovarian stromal tissue were significantly higher in postmenopausal vs. premenopausal women (mean ± SEM, 160.52 ± 17.75 vs. 120.67 ± 14.33; P=.002). Stronger Picrosirius Red staining, suggestive of fibrosis, was seen in ovarian stromal tissue of postmenopausal vs. premenopausal women (54.06 ± 6.94 vs. 37.50 ± 14.29; P=.02). Analysis of qPCR revealed that (1) Col1a1, Col1a2, Col3a1 levels were significantly lower in ovaries obtained from 14-month-old Fkbp5-/- vs. Fkbp5+/+ mice; (2) FKBP5 levels significantly increased in CCs of advanced age women vs. younger women (1.71 ± 0.22 vs. 1.11 ± 0.15; P=.03); and (3) FKBP5 levels were approximately threefold higher in GCs of women with DOR vs. age-matched control (3.22 ± 1.11 vs. 1.30 ± 0.54; P=.03). CONCLUSION This study for the first time demonstrates expression profile of FKBP51 in human ovary and its potential role in ovarian aging. Our results indicate that the up-regulation of FKBP51 is associated with ovarian aging. Moreover, in women undergoing in vitro fertilization treatment, enhanced FKBP51 expression is seen in those with DOR or women of advanced maternal reproductive age, who have poor prognosis. Therefore, drugs targeting inhibition of FKBP51 expression and/or activity may delay ovarian aging or treat premature ovarian aging.
Collapse
Affiliation(s)
- Papri Sarkar
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, Florida; Shady Grove Fertility, Tampa, Florida
| | - Monica Moore
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Asli Ozmen
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Busra Cetinkaya-Un
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Vitko Julie
- Department of Pathology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Anthony N Imudia
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, Florida; Shady Grove Fertility, Tampa, Florida
| | - Charles J Lockwood
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Umit A Kayisli
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Ozlem Guzeloglu-Kayisli
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, Florida.
| |
Collapse
|
25
|
Yoshino O, Ono Y. The physiological role of macrophages in reproductive organs. Reprod Med Biol 2025; 24:e12637. [PMID: 39959577 PMCID: PMC11827100 DOI: 10.1002/rmb2.12637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/04/2025] [Indexed: 02/18/2025] Open
Abstract
Background Macrophages are essential immune cells critical to reproductive physiology. They regulate key processes such as follicular development, ovulation, and luteinization in the ovaries. Macrophages are also involved in endometrial remodeling, immune tolerance, and placentation in the uterus. Methods This review examined the biological characteristics of macrophages and their role in ovarian, uterine, and fallopian tube physiology. It focused on findings from both animal and human studies to provide a comprehensive understanding of macrophage functions. Main Findings In the ovaries, M1 macrophages play a role in folliculogenesis and ovulation through the inflammatory and angiogenic pathways. Macrophages also maintain the corpus luteum and vascular integrity. In the uterus, macrophages regulate tissue repair and remodeling during the menstrual cycle and play a critical role in implantation by maintaining immune tolerance and supporting decidualization. Dysregulation of the M1/M2 balance can cause implantation failure. In the fallopian tubes, macrophages mediate tissue repair and immune responses. Macrophage polarization dynamically adapts to physiological and pathological conditions in all reproductive organs highlighting the functional plasticity of these cells. Conclusion Macrophage polarization and functions are pivotal in maintaining reproductive health. Hence, understanding the role of macrophages in various reproductive organs provides a foundation for developing new therapies.
Collapse
Affiliation(s)
- Osamu Yoshino
- Department of Obstetrics and GynecologyUniversity of YamanashiYamanashiJapan
| | - Yosuke Ono
- Department of Obstetrics and GynecologyUniversity of YamanashiYamanashiJapan
| |
Collapse
|
26
|
Isola JVV, Biswas S, Jayarathne H, Hubbart CR, Hense JD, Matsuzaki S, Kinter MT, Humphries KM, Ocañas SR, Sadagurski M, Stout MB. Canagliflozin treatment prevents follicular exhaustion and attenuates hallmarks of ovarian aging in genetically heterogenous mice. GeroScience 2024:10.1007/s11357-024-01465-w. [PMID: 39672978 DOI: 10.1007/s11357-024-01465-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: 09/21/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024] Open
Abstract
Ovarian aging is characterized by declines in follicular reserve and the emergence of mitochondrial dysfunction, reactive oxygen species production, inflammation, and fibrosis, which eventually results in menopause. Menopause is associated with increased systemic aging and the development of numerous comorbidities; therefore, the attenuation of ovarian aging could also delay systemic aging processes in women. Recent work has established that the anti-diabetic drug Canagliflozin (Cana), a sodium-glucose transporter 2 inhibitor, elicits benefits on aging-related outcomes, likely through the modulation of nutrient-sensing pathways and metabolic homeostasis. Given that nutrient-sensing pathways play a critical role in controlling primordial follicle activation, we sought to determine if chronic Cana administration would delay ovarian aging and curtail the emergence of pathological hallmarks associated with reproductive senescence. We found that mice receiving Cana maintained their ovarian reserve through 12 months of age, which was associated with declines in primordial follicles FoxO3a phosphorylation, a marker of activation, when compared to the age-matched controls. Furthermore, Cana treatment led to decreased collagen, lipofuscin, and T cell accumulation at 12 months of age. Whole ovary transcriptomic and proteomic analyses revealed subtle improvements, predominantly in mitochondrial function and the regulation of cellular proliferation. Pathway analyses of the transcriptomic data revealed a downregulation in cell proliferation and mitochondrial dysfunction signatures, with an upregulation of oxidative phosphorylation. Pathway analyses of the proteomic data revealed declines in signatures associated with PI3K/AKT activity and lymphocyte accumulation. Collectively, we demonstrate that Cana treatment can delay ovarian aging in mice and could potentially have efficacy for delaying ovarian aging in women.
Collapse
Affiliation(s)
- José V V Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Subhasri Biswas
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Hashan Jayarathne
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
| | - Chase R Hubbart
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Jessica D Hense
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Satoshi Matsuzaki
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Michael T Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Kenneth M Humphries
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA
| | - Sarah R Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Marianna Sadagurski
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13 Street, Chapman E306, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
27
|
Jiang W, Sun W, Peng Y, Xu H, Fan H, Jin X, Xiao Y, Wang Y, Yang P, Shu W, Li J. Single-cell RNA sequencing reveals the intercellular crosstalk and the regulatory landscape of stromal cells during the whole life of the mouse ovary. LIFE MEDICINE 2024; 3:lnae041. [PMID: 39872151 PMCID: PMC11748273 DOI: 10.1093/lifemedi/lnae041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 12/25/2024] [Indexed: 01/29/2025]
Abstract
The heterogeneity of ovarian mesenchymal/stromal cells has just been revealed in both mice and humans. However, it remains unclear about the cellular development trace and the intercellular communication network in the whole life of the ovary. In the study, we integrated ours and published single-cell RNA sequencing data from E11.5 (embryonic day 11.5) until M12 (12-month-old) ovaries to show the dynamics of somatic cells along the developmental timeline. The intercellular crosstalk among somatic cell types was depicted with collagen signaling pathway as the most outgoing signals from stromal cells. We identified mesenchymal progenitor cells (CD24+) as the origin of stromal cells. Although their numbers decreased significantly in adults, the cells served as the major signal sender until ovarian senescence. Moreover, the ovarian injury could activate these stem cells and induce stroma remodeling in the aged ovary. Thus, mesenchymal progenitor cells may represent a new strategy to delay ovarian aging in the future.
Collapse
Affiliation(s)
- Wan Jiang
- Bioinformatics Center of AMMS, Beijing 100850, China
| | - Wenya Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210029, China
| | - Yue Peng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210029, China
| | - Hao Xu
- Bioinformatics Center of AMMS, Beijing 100850, China
| | - Haonan Fan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210029, China
| | - Xin Jin
- Wuxi Maternity and Child Health Care Hospital, Affiliated Women’s Hospital of Jiangnan University, Wuxi 214000, China
| | - Yue Xiao
- The First Affiliated Hospital of Zhejiang University School of Medicine, Center of Reproductive Medicine, Hangzhou 310009, China
| | - Yuxiang Wang
- Bioinformatics Center of AMMS, Beijing 100850, China
| | - Pin Yang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230022, China
| | - Wenjie Shu
- Bioinformatics Center of AMMS, Beijing 100850, China
| | - Jing Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
28
|
Balough JL, Dipali SS, Velez K, Kumar TR, Duncan FE. Hallmarks of female reproductive aging in physiologic aging mice. NATURE AGING 2024; 4:1711-1730. [PMID: 39672896 DOI: 10.1038/s43587-024-00769-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/28/2024] [Indexed: 12/15/2024]
Abstract
The female reproductive axis is one of the first organ systems to age, which has consequences for fertility and overall health. Here, we provide a comprehensive overview of the biological process of female reproductive aging across reproductive organs, tissues and cells based on research with widely used physiologic aging mouse models, and describe the mechanisms that underpin these phenotypes. Overall, aging is associated with dysregulation of the hypothalamic-pituitary-ovarian axis, perturbations of the ovarian stroma, reduced egg quantity and quality, and altered uterine morphology and function that contributes to reduced capacity for fertilization and impaired embryo development. Ultimately, these age-related phenotypes contribute to altered pregnancy outcomes and adverse consequences in offspring. Conserved mechanisms of aging, as well as those unique to the reproductive system, underlie these phenotypes. The knowledge of such mechanisms will lead to development of therapeutics to extend female reproductive longevity and support endocrine function and overall health.
Collapse
Affiliation(s)
- Julia L Balough
- Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, USA
| | - Shweta S Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Karen Velez
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - T Rajendra Kumar
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Francesca E Duncan
- Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, USA.
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
29
|
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
|
30
|
Zheng M, Li J, Cao Y, Bao Z, Dong X, Zhang P, Yan J, Liu Y, Guo Y, Zeng X. Association of different inflammatory indices with risk of early natural menopause: a cross-sectional analysis of the NHANES 2013-2018. Front Med (Lausanne) 2024; 11:1490194. [PMID: 39678034 PMCID: PMC11638831 DOI: 10.3389/fmed.2024.1490194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Background Early natural menopause, characterized by the cessation of ovarian function before the age of 45, has been a subject of prior research indicating that inflammation may predict the onset of menopause. However, the specific relationship between peripheral blood inflammatory parameters and early natural menopause remains ambiguous. Methods This observational study utilized data from the National Health and Nutrition Examination Survey (NHANES) spanning the years 2013-2018. The age at menopause was ascertained through the Reproductive Health Questionnaire (RHQ), with early natural menopause defined as menopause occurring before the age of 45 years. Complete blood counts were derived from laboratory test data, and seven indices of inflammation were calculated, including lymphocyte count (LC), neutrophil count (NC), systemic immune inflammation index (SII), product of platelet and neutrophil count (PPN), platelet-lymphocyte ratio (PLR), neutrophil-lymphocyte ratio (NLR), and lymphocyte-monocyte ratio (LMR). A multivariate weighted logistic regression analysis was employed to estimate the association between these inflammatory indices and early natural menopause. Results A total of 2,034 participants were included in the analysis, of whom 460 reported experiencing menopause before the age of 45. Both Log2-NC and Log2-PPN were found to be positively correlated with early menopause, with odds ratios (OR) of 1.56 (95% CI: 1.16, 2.09; p = 0.005) and 1.36 (95% CI: 1.07, 1.72; p = 0.015), respectively. The results from models 1 and 2 were consistent with those from model 3. In the trend test, participants in the fourth quartile (Q4) of log2-LC exhibited a positive correlation with early menopause compared to those in the lowest quartile (Q1), with an OR of 1.41 (95% CI: 1.03, 1.93; p = 0.033). Similarly, the fourth quartile (Q4) of log2-NC and log2-PPN demonstrated a positive correlation with early menopause, with odds ratios (OR) of 1.76 (95% CI: 1.27-2.45; p < 0.001) and 1.66 (95% CI: 1.21-2.29; p = 0.002), respectively. In Model 3, log2-SII, log2-PLR, log2-NLR, and log2-LMR were not significantly associated with early menopause. Conclusion Our findings indicate that elevated levels of Log2-LC, Log2-NC, and Log2-PPN are positively correlated with an increased risk of early menopause among women in the United States.
Collapse
Affiliation(s)
- Mengyu Zheng
- Department of Gynecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
| | - Junying Li
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yushan Cao
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhuo Bao
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xing Dong
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pei Zhang
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinxiang Yan
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yixuan Liu
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongzhen Guo
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianxu Zeng
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
31
|
Guo H, Xiao C, Li X, Li J, Chen X, Bin Liu, Hu R. PAI-1 siRNA-loaded biomimetic nanoparticles for ameliorating diminished ovarian reserve and inhibiting ovarian fibrosis. Eur J Pharmacol 2024; 983:176948. [PMID: 39216744 DOI: 10.1016/j.ejphar.2024.176948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
With specific and inherent mRNA cleaving activity, small interfering RNA against pro-fibrosis factor (PAI-1 siRNA, siPAI-1) has demonstrated the fucntion for preventing diminished ovarian reserve (DOR). Moreover, safe nanomaterials have provided ideal tools for delivering siRNA to the targeted cells to obtain high therapeutic efficacy. In order to improve the preventing capability of siPAI-1 for DOR, we synthesized one kind of biomimetic Poly (lactic-co-glycolic acid) copolymer (PLGA)-based nanoparticles (siPAI-1@PLGA@M-FSHL, abbreviated as SPMF). siPAI-1 was assembled into cationic PLGA nanoparticles, following with macrophage membrane coating (M) and FSHL81-95 peptide modification. SPMF NPs significantly enhanced cellular uptake and gene silencing efficiency in KGN cells in vitro. In vivo assay demonstrated that SPMF NPs can targetedly accumulate in the ovarian of DOR mice with Cyclophosphamide treatment (80 mg/kg/week, 2 weeks) and remarkably downregulate the levels of PAI-1 in ovarian, which finally resulted in the effective suppression of ovary fibrosis and improved the chemotherapy-induced follicle loss to increase the number of primordial, secondary, antral follicles by 62.05 %, 54.92 % and 64.37 %, respectively, compared with DOR group. In summary, this study demonstrates that siPAI-1-loaded SPMF with high safety and efficacy can potentially alleviate DOR by inhibiting the overexpression of PAI-1 in the ovarian.
Collapse
Affiliation(s)
- Hua Guo
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China; Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Chang Xiao
- College of Biology, Hunan University, Changsha, 410082, China
| | - Xinshu Li
- Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Jialing Li
- Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Xue Chen
- Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, China.
| | - Rong Hu
- Reproductive Medicine Center, General Hosptial of Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
32
|
Yan L, Tu W, Zhao X, Wan H, Wu J, Zhao Y, Wu J, Sun Y, Zhu L, Qin Y, Hu L, Yang H, Ke Q, Zhang W, Luo W, Xiao Z, Chen X, Wu Q, He B, Teng M, Dai S, Zhai J, Wu H, Yang X, Guo F, Wang H. Stem cell transplantation extends the reproductive life span of naturally aging cynomolgus monkeys. Cell Discov 2024; 10:111. [PMID: 39496598 PMCID: PMC11535534 DOI: 10.1038/s41421-024-00726-4] [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: 12/18/2023] [Accepted: 08/03/2024] [Indexed: 11/06/2024] Open
Abstract
The ovary is crucial for female reproduction and health, as it generates oocytes and secretes sex hormones. Transplantation of mesenchymal stem cells (MSCs) has been shown to alleviate pathological ovarian aging. However, it is unclear whether MSCs could benefit the naturally aging ovary. In this study, we first examined the dynamics of ovarian reserve of Chinese women during perimenopause. Using a naturally aging cynomolgus monkey (Macaca fascicularis) model, we found that transplanting human embryonic stem cells-derived MSC-like cells, which we called M cells, into the aging ovaries significantly decreased ovarian fibrosis and DNA damage, enhanced secretion of sex hormones and improved fertility. Encouragingly, a healthy baby monkey was born after M-cell transplantation. Moreover, single-cell RNA sequencing analysis and in vitro functional validation suggested that apoptosis, oxidative damage, inflammation, and fibrosis were mitigated in granulosa cells and stromal cells following M-cell transplantation. Altogether, these findings demonstrate the beneficial effects of M-cell transplantation on aging ovaries and expand our understanding of the molecular mechanisms underlying ovarian aging and stem cell-based alleviation of this process.
Collapse
Affiliation(s)
- Long Yan
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Wan Tu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Xuehan Zhao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Haifeng Wan
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jiaqi Wu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yan Zhao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jun Wu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
| | - Yingpu Sun
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lan Zhu
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yingying Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
| | - Linli Hu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hua Yang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qiong Ke
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenzhe Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wei Luo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhenyu Xiao
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xueyu Chen
- Laboratory of Neonatology, Department of Neonatology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Qiqian Wu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Beijia He
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Man Teng
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Shanjun Dai
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jinglei Zhai
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Hao Wu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Xiaokui Yang
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fan Guo
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Hongmei Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| |
Collapse
|
33
|
Xia X, Yang Y, Liu P, Chen L, Dai X, Xue P, Wang Y. The senolytic drug ABT-263 accelerates ovarian aging in older female mice. Sci Rep 2024; 14:23178. [PMID: 39369073 PMCID: PMC11457520 DOI: 10.1038/s41598-024-73828-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
Previous studies have reported that senolytic drugs can reverse obesity-mediated accumulation of senescent cells in the ovary and protect against cisplatin-induced ovarian injury by removing senescent cells. Early intervention with ABT-263 has been shown to mitigate ovarian aging. However, it remains unknown whether treatment with ABT-263 could rejuvenate the aged ovary in reproductively old females. Therefore, the current study was aimed to investigate whether advanced age intervention with ABT-263 could ameliorate age-related decline in ovarian function. Fourteen 16-month-old mice with a C57/BL6 background were treated with ABT-263 (N = 7) or vehicle (N = 7) for two weeks. Mice were initially treated with ABT-263 (60 mg/kg/d) or vehicle for 7 consecutive days. After a 7-day break, the treatment was repeated for another 7 consecutive days. Six 2-month-old mice with C57BL/6 were used as a young control. The hormonal levels, estrus cycles, ovarian reserve, ovarian cell proliferation and apoptosis, ovarian fibrosis, and steroidogenic gene expression of ovarian stromal cells were evaluated. ABT-263 treatment did not rescue abnormal estrus cycles and sex hormonal levels, or inhibit the formation of multinucleated giant cells and ovarian stromal cell apoptosis in aged ovaries. However, it reduced ovarian fibrosis and preserved the steroidogenic gene expression of ovarian stromal cells in aged ovaries. Importantly, ABT-263 treatment further depleted ovarian follicles in aged mice. In conclusion, ABT-263 treatment accelerated the depletion of ovarian follicles in aged mice, suggesting that senolytic drugs for reproductively old female may adversely affect female fertility.
Collapse
Affiliation(s)
- Xiyang Xia
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yingying Yang
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Pengfei Liu
- The Department of Animal Center, Kebiao Medical Testing Center, Changzhou, Jiangsu, China
| | - Li Chen
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Xiuliang Dai
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China.
| | - Pingping Xue
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China.
| | - Yufeng Wang
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China.
| |
Collapse
|
34
|
Yao L, Yang C, Graff JC, Wang G, Wang G, Gu W. From Reactive to Proactive - The Future Life Design to Promote Health and Extend the Human Lifespan. Adv Biol (Weinh) 2024; 8:e2400148. [PMID: 39037380 DOI: 10.1002/adbi.202400148] [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: 03/15/2024] [Revised: 06/11/2024] [Indexed: 07/23/2024]
Abstract
Disease treatment and prevention have improved the human lifespan. Current studies on aging, such as the biological clock and senolytic drugs have focused on the medical treatments of various disorders and health maintenance. However, to efficiently extend the human lifespan to its theoretical maximum, medicine can take a further proactive approach and identify the inapparent disorders that affect the gestation, body growth, and reproductive stages of the so-called "healthy" population. The goal is to upgrade the standard health status to a new level by targeting the inapparent disorders. Thus, future research can shift from reaction, response, and prevention to proactive, quality promotion and vigor prolonging; from single disease-oriented to multiple dimension protocol for a healthy body; from treatment of symptom onset to keep away from disorders; and from the healthy aging management to a healthy promotion design beginning at the birth.
Collapse
Affiliation(s)
- Lan Yao
- College of Health management, Harbin Medical University, 157 Baojian Road, Harbin, Heilongjiang, 150081, China
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Chengyuan Yang
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - J Carolyn Graff
- Department of Health Promotion and Disease Prevention, College of Nursing, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Guiying Wang
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150007, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150007, China
| | - Weikuan Gu
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
- Research Service, Memphis VA Medical Center, 1030 Jefferson Avenue, Memphis, TN, 38104, USA
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN, 38163, USA
| |
Collapse
|
35
|
Pereira LAAC, Ferreira CS, Dias KSSA, Nogueira JM, Pinto FCH, Jorge EC, Campos-Junior PHA. Ovarian Puncture Triggers an Inflammatory Response that did not Affect Late Folliculogenesis, Ovulation Rate, and Fertility. Reprod Sci 2024; 31:3202-3214. [PMID: 39043998 DOI: 10.1007/s43032-024-01654-0] [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: 10/17/2023] [Accepted: 07/11/2024] [Indexed: 07/25/2024]
Abstract
Ovarian puncture has been widely used in assisted reproduction, but there are still gaps about its effects on ovarian morphophysiology, as well as the relationship between inflammation caused by this procedure and the follicular growth and fertility. The aim of this study was to investigate the effects of ovarian puncture on folliculogenesis and fertility. Mice (n = 24) were divided into two groups: (1) SHAM-both ovaries were exposed and repositioned and (2) Punctured-ovaries were exposed, punctured, and repositioned. After 96 h of surgery, ovaries were collected for morphofunctional analysis. New females were used for the superovulation (n = 10) and fertility assays (n = 10). Increased volumetric density of inflammatory cells-p = 0.0005, p = 0.0013; hemorrhagic foci-p < 0.0001; and inflammatory exudate-p < 0.0001 could be noticed on the punctured group, compared to SHAM. The percentage of primordial follicles was lower on the punctured ovaries (p = 0.00294). Ovarian puncture has also induced an increase in the proliferation of granulosa cells of primary (p = 0.0321) and antral follicles (p = 0.0395), and an increased apoptotic index of antral follicles (p = 0.0100). There was no influence on expression of some genes related to inflammation, collagen deposition and folliculogenesis progression. The reproductive aspects (oocyte retrieval and number of fetuses per female) were not altered (p > 0.05). Taken together, our findings strongly suggest that ovarian puncture results in a local inflammation that affects follicular growth and atresia. However, it does not affect female fertility, which strengthens the safety of this procedure.
Collapse
Affiliation(s)
| | - Camila Stefane Ferreira
- Laboratory for Reproductive Biology Research, Department of Natural Sciences, Federal University of São João del Rei, São João Del-Rei, MG, Brazil
| | - Karine Sthéfany Serpa Amaral Dias
- Laboratory for Reproductive Biology Research, Department of Natural Sciences, Federal University of São João del Rei, São João Del-Rei, MG, Brazil
| | - Júlia Meireles Nogueira
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Flávia Carmo Horta Pinto
- Laboratory for Reproductive Biology Research, Department of Natural Sciences, Federal University of São João del Rei, São João Del-Rei, MG, Brazil
| | - Erika Cristina Jorge
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Paulo Henrique Almeida Campos-Junior
- Laboratory for Reproductive Biology Research, Department of Natural Sciences, Federal University of São João del Rei, São João Del-Rei, MG, Brazil.
| |
Collapse
|
36
|
Liang J, Huang F, Hao X, Zhang P, Chen R. Nicotinamide mononucleotide supplementation rescues mitochondrial and energy metabolism functions and ameliorates inflammatory states in the ovaries of aging mice. MedComm (Beijing) 2024; 5:e727. [PMID: 39355508 PMCID: PMC11442848 DOI: 10.1002/mco2.727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 10/03/2024] Open
Abstract
Noninvasive pharmacological strategies like nicotinamide mononucleotide (NMN) supplementation can effectively address age-related ovarian infertility by maintaining or enhancing oocyte quality and quantity. This study revealed that ovarian nicotinamide adenine dinucleotide levels decline with age, but NMN administration significantly restores these levels, preventing ovarian atrophy and enhancing the quality and quantity of ovulated oocytes. Improvements in serum hormone secretion and antioxidant factors, along with decreased expression of proinflammatory factors, were observed. Additionally, a significant increase in the number of ovarian follicles in aging individuals was noted. Scanning electron microscopy data indicated that NMN significantly alters the density and morphology of lipid droplets and mitochondria in granulosa cells, suggesting potential targets and mechanisms. Transcriptomic analysis and validation experiments collectively suggested that the beneficial effects of NMN on aging ovaries are mediated through enhanced mitochondrial function, improved energy metabolism, and reduced inflammation levels. Our results suggest that NMN supplementation could improve the health status of aging ovaries and enhance ovarian reserve, offering new insights into addressing fertility challenges in older women through assisted reproductive technology.
Collapse
Affiliation(s)
- Jinghui Liang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College National Clinical Research Center for Obstetric & Gynecologic Diseases Beijing China
| | - Feiling Huang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College National Clinical Research Center for Obstetric & Gynecologic Diseases Beijing China
| | - Xueyu Hao
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University National Center for Children's Health Beijing China
- MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University National Center for Children's Health Beijing China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Rare Disease Center, Beijing Children's Hospital, Capital Medical University National Center for Children's Health Beijing China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University National Center for Children's Health Beijing China
- MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University National Center for Children's Health Beijing China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Rare Disease Center, Beijing Children's Hospital, Capital Medical University National Center for Children's Health Beijing China
| | - Rong Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College National Clinical Research Center for Obstetric & Gynecologic Diseases Beijing China
| |
Collapse
|
37
|
Amargant F, Magalhaes C, Pritchard MT, Duncan FE. Systemic low-dose anti-fibrotic treatment attenuates ovarian aging in the mouse. GeroScience 2024:10.1007/s11357-024-01322-w. [PMID: 39285140 DOI: 10.1007/s11357-024-01322-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: 06/24/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
The female reproductive system is one of the first to age in humans, resulting in infertility and endocrine disruptions. The aging ovary assumes a fibro-inflammatory milieu which negatively impacts gamete quantity and quality as well as ovulation. Here, we tested whether the systemic delivery of anti-inflammatory (Etanercept) or anti-fibrotic (Pirfenidone) drugs attenuates ovarian aging in mice. We first evaluated the ability of these drugs to decrease the expression of fibro-inflammatory genes in primary ovarian stromal cells treated with a pro-fibrotic or a pro-inflammatory stimulus. Whereas Etanercept did not block Tnf expression in ovarian stromal cells, Pirfenidone significantly reduced Col1a1 expression. We then tested Pirfenidone in vivo where the drug was delivered systemically via mini-osmotic pumps for 6 weeks. Pirfenidone mitigated the age-dependent increase in ovarian fibrosis without impacting overall health parameters. Ovarian function was improved in Pirfenidone-treated mice as evidenced by increased follicle and corpora lutea number, AMH levels, and improved estrous cyclicity. Transcriptomic analysis revealed that Pirfenidone treatment resulted in an upregulation of reproductive function-related genes at 8.5 months and a downregulation of inflammatory genes at 12 months of age. These findings demonstrate that reducing the fibroinflammatory ovarian microenvironment improves ovarian function, thereby supporting modulating the ovarian environment as a therapeutic avenue to extend reproductive longevity.
Collapse
Affiliation(s)
- Farners Amargant
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Carol Magalhaes
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michele T Pritchard
- Department of Pharmacology, Toxicology and Therapeutics and Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
38
|
Di Emidio G, Vergara T, Konstantinidou F, Flati I, Stuppia L, Artini PG, Gatta V, Falone S, Tatone C. NAD + Metabolism and Mitochondrial Activity in the Aged Oocyte: Focus on the Effects of NAMPT Stimulation. Aging Dis 2024; 15:2828-2851. [PMID: 39325941 PMCID: PMC11567263 DOI: 10.14336/ad.2024.0241] [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: 03/12/2024] [Accepted: 07/16/2024] [Indexed: 09/28/2024] Open
Abstract
The ovary experiences an age-dependent decline starting during the fourth decade of life. Ovarian aging is the predominant factor driving female reproductive aging. Modern trend to postpone childbearing age contributes to reduced fertility and natality worldwide. Recently, the beneficial role of NAD+ precursors on the maintenance of oocyte competence and female fertility affected by aging has emerged. Nevertheless, age-related changes in NAD+ regulatory network have not been investigated so far. In this context, our goal was to investigate changes induced by the aging process in the expression level of genes participating in NAD+ biosynthetic and NAD+ consuming pathways and in the cellular bioenergetics in the mouse oocyte. From Ingenuity Pathway Analysis (IPA) it emerged that aging caused the downregulation of all cellular pathways for NAD+ synthesis (Kynurenine pathway, Preiss-Handler pathway and NAD+ salvage pathway) and deeply influenced the activity of NAD+-dependent enzymes, i.e. PARPs and SIRTs, with effects on many cellular functions including compromised ROS detoxification. Considering that NAMPT, the rate-limiting enzyme of NAD+ salvage pathway, was deregulated, aged oocytes were matured in the presence of P7C3, NAMPT activator. P7C3 improved spindle assembly and mitochondrial bioenergetics and reduced mitochondrial proton leak. Moreover, P7C3 influenced gene expression of NAD+ regulatory network, with Sirt1 as the central node of IPA-interfered target gene network. Finally, P7C3 effectively counteracted oocyte alterations induced by exposure to oxidative stress. Our study contributes to establish effective NAD+ boosting interventions to alleviate the effects of advanced maternal age on fertility and explore their potential in redox-related fertility disorders.
Collapse
Affiliation(s)
- Giovanna Di Emidio
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy.
| | - Teresa Vergara
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy.
| | - Fani Konstantinidou
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy.
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy.
| | - Irene Flati
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy.
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy.
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy.
| | - Paolo Giovanni Artini
- Department of Obstetrics and Gynecology “P. Fioretti”, University of Pisa, 56126 Pisa, Italy.
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy.
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy.
| | - Stefano Falone
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy.
| | - Carla Tatone
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy.
| |
Collapse
|
39
|
Morimoto A, Rose RD, Smith KM, Dinh DT, Umehara T, Winstanley YE, Shibahara H, Russell DL, Robker RL. Granulosa cell metabolism at ovulation correlates with oocyte competence and is disrupted by obesity and aging. Hum Reprod 2024; 39:2053-2066. [PMID: 39013118 PMCID: PMC11373349 DOI: 10.1093/humrep/deae154] [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: 11/18/2023] [Revised: 06/16/2024] [Indexed: 07/18/2024] Open
Abstract
STUDY QUESTION Is oocyte developmental competence associated with changes in granulosa cell (GC) metabolism? SUMMARY ANSWER GC metabolism is regulated by the LH surge, altered by obesity and reproductive aging, and, in women, specific metabolic profiles are associated with failed fertilization versus increased blastocyst development. WHAT IS KNOWN ALREADY The cellular environment in which an oocyte matures is critical to its future developmental competence. Metabolism is emerging as a potentially important factor; however, relative energy production profiles between GCs and cumulus cells and their use of differential substrates under normal in vivo ovulatory conditions are not well understood. STUDY DESIGN, SIZE, DURATION This study identified metabolic and substrate utilization profiles within ovarian cells in response to the LH surge, using mouse models and GCs of women undergoing gonadotropin-induced oocyte aspiration followed by IVF/ICSI. PARTICIPANTS/MATERIALS, SETTING, METHODS To comprehensively assess follicular energy metabolism, we used real-time metabolic analysis (Seahorse XFe96) to map energy metabolism dynamics (mitochondrial respiration, glycolysis, and fatty acid oxidation) in mouse GCs and cumulus-oocyte complexes (COCs) across a detailed time course in the lead up to ovulation. In parallel, the metabolic profile of GCs was measured in a cohort of 85 women undergoing IVF/ICSI (n = 21 with normal ovarian function; n = 64 with ovarian infertility) and correlated with clinical parameters and cycle outcomes. MAIN RESULTS AND THE ROLE OF CHANCE Our study reveals dynamic changes in GC energy metabolism in response to ovulatory LH, with mitochondrial respiration and glycolysis differentially affected by obesity versus aging, in both mice and women. High respiration in GCs is associated with failed fertilization (P < 0.05) in a subset of women, while glycolytic reserve and mitochondrial ATP production are correlated with on-time development at Day 3 (P < 0.05) and blastocyst formation (P < 0.01) respectively. These data provide new insights into the cellular mechanisms of infertility, by uncovering significant associations between metabolism within the ovarian follicle and oocyte developmental competence. LIMITATIONS, REASONS FOR CAUTION A larger prospective study is needed before the metabolic markers that were positively and negatively associated with oocyte quality can be used clinically to predict embryo outcomes. WIDER IMPLICATIONS OF THE FINDINGS This study offers new insights into the importance of GC metabolism for subsequent embryonic development and highlights the potential for therapeutic strategies focused on optimizing mitochondrial metabolism to support embryonic development. STUDY FUNDING/COMPETING INTEREST(S) National Health and Medical Research Council (Australia). The authors have no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Atsushi Morimoto
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
- Department of Obstetrics and Gynecology, School of Medicine, Hyogo Medical University, Hyogo, Japan
| | - Ryan D Rose
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
- Genea Fertility SA, Adelaide, SA, Australia
| | - Kirsten M Smith
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Doan T Dinh
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Takashi Umehara
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Yasmyn E Winstanley
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Hiroaki Shibahara
- Department of Obstetrics and Gynecology, School of Medicine, Hyogo Medical University, Hyogo, Japan
| | - Darryl L Russell
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Rebecca L Robker
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
40
|
Yan Y, Zhang H, Xu R, Luo L, Yin L, Wu H, Zhang Y, Li C, Lu S, Tang Y, Zhao X, Pan M, Wei Q, Peng S, Ma B. Single-cell sequencing reveals the transcriptional alternations of 17β-estradiol suppressing primordial follicle formation in neonatal mouse ovaries. Cell Prolif 2024; 57:e13713. [PMID: 38988058 PMCID: PMC11503257 DOI: 10.1111/cpr.13713] [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: 02/22/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
Estrogen has been implicated in multiple biological processes, but the variation underlying estrogen-mediated primordial follicle (PF) formation remains unclear. Here, we show that 17β-estradiol (E2) treatment of neonatal mice led to the inhibition of PF formation and cell proliferation. Single-cell RNA sequencing (scRNA-seq) revealed that E2 treatment caused significant changes in the transcriptome of oocytes and somatic cells. E2 treatment disrupted the synchronised development of oocytes, pre-granulosa (PG) cells and stromal cells. Mechanistically, E2 treatment disrupted several signalling pathways critical to PF formation, especially down-regulating the Kitl and Smad1/3/4/5/7 expression, reducing the frequency and number of cell communication. In addition, E2 treatment influenced key gene expression, mitochondrial function of oocytes, the recruitment and maintenance of PG cells, the cell proliferation of somatic cells, as well as disordered the ovarian microenvironment. This study not only revealed insights into the regulatory role of estrogen during PF formation, but also filled in knowledge of dramatic changes in perinatal hormones, which are critical for the physiological significance of understanding hormone changes and reproductive protection.
Collapse
Affiliation(s)
- Yutong Yan
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Hui Zhang
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Rui Xu
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Linglin Luo
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Lu Yin
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Hao Wu
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Yiqian Zhang
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Chan Li
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Sihai Lu
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Yaju Tang
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Xiaoe Zhao
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Menghao Pan
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Qiang Wei
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Sha Peng
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Baohua Ma
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| |
Collapse
|
41
|
Dipali SS, Gowett MQ, Kamat P, Converse A, Zaniker EJ, Fennell A, Chou T, Pritchard MT, Zelinski M, Phillip JM, Duncan FE. Self-organizing ovarian somatic organoids preserve cellular heterogeneity and reveal cellular contributions to ovarian aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.10.607456. [PMID: 39211064 PMCID: PMC11360955 DOI: 10.1101/2024.08.10.607456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Ovarian somatic cells are essential for reproductive function, but no existing ex vivo models recapitulate the cellular heterogeneity or interactions within this compartment. We engineered a novel ovarian somatic organoid model by culturing a stroma-enriched fraction of mouse ovaries in scaffold-free agarose micromolds. Ovarian somatic organoids self-organized, maintained diverse cell populations, produced extracellular matrix, and secreted hormones. Organoids generated from reproductively old mice exhibited reduced aggregation and growth compared to young counterparts, as well as differences in cellular composition. Interestingly, matrix fibroblasts from old mice demonstrated upregulation of pathways associated with the actin cytoskeleton and downregulation of cell adhesion pathways, indicative of increased cellular stiffness which may impair organoid aggregation. Cellular morphology, which is regulated by the cytoskeleton, significantly changed with age and in response to actin depolymerization. Moreover, actin depolymerization rescued age-associated organoid aggregation deficiency. Overall, ovarian somatic organoids have advanced fundamental knowledge of cellular contributions to ovarian aging.
Collapse
|
42
|
Gokyer D, Akinboro S, Zhou LT, Kleinhans A, Laronda MM, Duncan FE, Riley JK, Goldman KN, Babayev E. The oocyte microenvironment is altered in adolescents compared to oocyte donors. Hum Reprod Open 2024; 2024:hoae047. [PMID: 39211054 PMCID: PMC11361810 DOI: 10.1093/hropen/hoae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/03/2024] [Indexed: 09/04/2024] Open
Abstract
STUDY QUESTION Do the molecular signatures of cumulus cells (CCs) and follicular fluid (FF) of adolescents undergoing fertility preservation differ from that of oocyte donors? SUMMARY ANSWER The microenvironment immediately surrounding the oocyte, including the CCs and FF, is altered in adolescents undergoing fertility preservation compared to oocyte donors. WHAT IS KNOWN ALREADY Adolescents experience a period of subfecundity following menarche. Recent evidence suggests that this may be at least partially due to increased oocyte aneuploidy. Reproductive juvenescence in mammals is associated with suboptimal oocyte quality. STUDY DESIGN SIZE DURATION This was a prospective cohort study. Adolescents (10-19 years old, n = 23) and oocyte donors (22-30 years old, n = 31) undergoing ovarian stimulation and oocyte retrieval at a single center between 1 November 2020 and 1 May 2023 were enrolled in this study. PARTICIPANTS/MATERIALS SETTING METHODS Patient demographics, ovarian stimulation, and oocyte retrieval outcomes were collected for all participants. The transcriptome of CCs associated with mature oocytes was compared between adolescents (10-19 years old, n = 19) and oocyte donors (22-30 years old, n = 19) using bulk RNA-sequencing. FF cytokine profiles (10-19 years old, n = 18 vs 25-30 years old, n = 16) were compared using cytokine arrays. MAIN RESULTS AND THE ROLE OF CHANCE RNA-seq analysis revealed 581 differentially expressed genes in CCs of adolescents relative to oocyte donors, with 361 genes downregulated and 220 upregulated. Genes enriched in pathways involved in cell cycle and cell division (e.g. GO: 1903047, P = 3.5 × 10-43; GO: 0051983, P = 4.1 × 10-30; GO: 0000281, P = 7.7 × 10-15; GO: 0044839, P = 5.3 × 10-13) were significantly downregulated, while genes enriched in several pathways involved in cellular and vesicle organization (e.g. GO: 0010256, P = 1.2 × 10-8; GO: 0051129, P = 6.8 × 10-7; GO: 0016050, P = 7.4 × 10-7; GO: 0051640, P = 8.1 × 10-7) were upregulated in CCs of adolescents compared to oocyte donors. The levels of nine cytokines were significantly increased in FF of adolescents compared to oocyte donors: IL-1 alpha (2-fold), IL-1 beta (1.7-fold), I-309 (2-fold), IL-15 (1.6-fold), TARC (1.9-fold), TPO (2.1-fold), IGFBP-4 (2-fold), IL-12-p40 (1.7-fold), and ENA-78 (1.4-fold). Interestingly, seven of these cytokines have known pro-inflammatory roles. Importantly, neither the CC transcriptomes nor FF cytokine profiles were different in adolescents with or without cancer. LARGE SCALE DATA Original high-throughput sequencing data have been deposited in Gene Expression Omnibus (GEO) database with the accession number GSE265995. LIMITATIONS REASONS FOR CAUTION This study aims to gain insights into the associated gamete quality by studying the immediate oocyte microenvironment. The direct study of oocytes is more challenging due to sample scarcity, as they are cryopreserved for future use, but would provide a more accurate assessment of oocyte reproductive potential. WIDER IMPLICATIONS OF THE FINDINGS Our findings have implications for the adolescent fertility preservation cycles. Understanding the expected quality of cryopreserved eggs in this age group will lead to better counseling of these patients about their reproductive potential and may help to determine the number of eggs that is recommended to be banked to achieve a reasonable chance of future live birth(s). STUDY FUNDING/COMPETING INTERESTS This project was supported by Friends of Prentice organization SP0061324 (M.M.L. and E.B.), Gesualdo Family Foundation (Research Scholar: M.M.L.), and NIH/NICHD K12 HD050121 (E.B.). The authors have declared that no conflict of interest exists.
Collapse
Affiliation(s)
- Dilan Gokyer
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sophia Akinboro
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neuroscience, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Luhan T Zhou
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anna Kleinhans
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, USA
| | - Monica M Laronda
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Basic and Preclinical Science, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joan K Riley
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, USA
| | - Kara N Goldman
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, USA
| | - Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, USA
| |
Collapse
|
43
|
Mihalas BP, Marston AL, Wu LE, Gilchrist RB. Reproductive Ageing: Metabolic contribution to age-related chromosome missegregation in mammalian oocytes. Reproduction 2024; 168:e230510. [PMID: 38718822 PMCID: PMC11301428 DOI: 10.1530/rep-23-0510] [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: 12/28/2023] [Accepted: 05/07/2024] [Indexed: 06/29/2024]
Abstract
In brief Chromosome missegregation and declining energy metabolism are considered to be unrelated features of oocyte ageing that contribute to poor reproductive outcomes. Given the bioenergetic cost of chromosome segregation, we propose here that altered energy metabolism during ageing may be an underlying cause of age-related chromosome missegregation and aneuploidy. Abstract Advanced reproductive age in women is a major cause of infertility, miscarriage and congenital abnormalities. This is principally caused by a decrease in oocyte quality and developmental competence with age. Oocyte ageing is characterised by an increase in chromosome missegregation and aneuploidy. However, the underlying mechanisms of age-related aneuploidy have not been fully elucidated and are still under active investigation. In addition to chromosome missegregation, oocyte ageing is also accompanied by metabolic dysfunction. In this review, we integrate old and new perspectives on oocyte ageing, chromosome segregation and metabolism in mammalian oocytes and make direct links between these processes. We consider age-related alterations to chromosome segregation machinery, including the loss of cohesion, microtubule stability and the integrity of the spindle assembly checkpoint. We focus on how metabolic dysfunction in the ageing oocyte disrupts chromosome segregation machinery to contribute to and exacerbate age-related aneuploidy. More specifically, we discuss how mitochondrial function, ATP production and the generation of free radicals are altered during ageing. We also explore recent developments in oocyte metabolic ageing, including altered redox reactions (NAD+ metabolism) and the interactions between oocytes and their somatic nurse cells. Throughout the review, we integrate the mechanisms by which changes in oocyte metabolism influence age-related chromosome missegregation.
Collapse
Affiliation(s)
- Bettina P Mihalas
- Oocyte Biology Research Unit, Discipline of Women’s Health, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Australia
| | - Adele L Marston
- Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Lindsay E Wu
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Kensington, Australia
| | - Robert B Gilchrist
- Oocyte Biology Research Unit, Discipline of Women’s Health, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Australia
| |
Collapse
|
44
|
Zeng Y, Wang C, Yang C, Shan X, Meng XQ, Zhang M. Unveiling the role of chronic inflammation in ovarian aging: insights into mechanisms and clinical implications. Hum Reprod 2024; 39:1599-1607. [PMID: 38906835 DOI: 10.1093/humrep/deae132] [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: 11/26/2023] [Revised: 05/14/2024] [Indexed: 06/23/2024] Open
Abstract
Ovarian aging, a natural process in women and various other female mammals as they age, is characterized by a decline in ovarian function and fertility due to a reduction in oocyte reserve and quality. This phenomenon is believed to result from a combination of genetic, hormonal, and environmental factors. While these factors collectively contribute to the shaping of ovarian aging, the substantial impact and intricate interplay of chronic inflammation in this process have been somewhat overlooked in discussions. Chronic inflammation, a prolonged and sustained inflammatory response persisting over an extended period, can exert detrimental effects on tissues and organs. This review delves into the novel hallmark of aging-chronic inflammation-to further emphasize the primary characteristics of ovarian aging. It endeavors to explore not only the clinical symptoms but also the underlying mechanisms associated with this complex process. By shining a spotlight on chronic inflammation, the aim is to broaden our understanding of the multifaceted aspects of ovarian aging and its potential clinical implications.
Collapse
Affiliation(s)
- Yutian Zeng
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Chun Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Cuiting Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Xudong Shan
- Genital Medicine Center, The Third People's Hospital of Cheng, Sichuan, China
| | - Xiang-Qian Meng
- Department of Reproductive Medicine, Sichuan Jinxin Xinan Woman & Children Hospital, Chengdu, China
| | - Ming Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
45
|
Yao Y, Wang B, Yu K, Song J, Wang L, Zhang X, Li Y. Nur77 improves ovarian function in reproductive aging mice by activating mitophagy and inhibiting apoptosis. Reprod Biol Endocrinol 2024; 22:86. [PMID: 39044215 PMCID: PMC11265396 DOI: 10.1186/s12958-024-01250-6] [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: 05/15/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Reproductive aging not only affects the fertility and physical and mental health of women but also accelerates the aging process of other organs. There is an urgent need newfor novel mechanisms, targets, and drugs to break the vicious cycle of mitochondrial dysfunction, redox imbalance, and germ cell apoptosis associated with ovarian aging. Autophagy, recognized as a longevity mechanism, has recently become a focal point in anti-aging research. Although mitophagy is a type of autophagy, its role and regulatory mechanisms in ovarian aging, particularly in age-related ovarian function decline, remain unclear. Nerve growth factor inducible gene B (Nur77) is an early response gene that can be stimulated by oxidative stress, DNA damage, metabolism, and inflammation. Recent evidence recommends that decreased expression of Nur77 is associated with age-related myocardial fibrosis, renal dysfunction, and Parkinson's disease; however, its association with ovarian aging has not been studied yet. We herein identified Nur77 as a regulator of germ cell senescence, apoptosis, and mitophagy and found that overexpression of Nur77 can activate mitophagy, improve oxidative stress, reduce apoptosis, and ultimately enhance ovarian reserve in aged mice ovaries. Furthermore, we discovered an association between Nur77 and the AKT pathway through String and molecular docking analyses. Experimental confirmation revealed that the AKT/mTOR signaling pathway is involved in the regulation of Nur77 in ovarian function. In conclusion, our results suggest Nur77 as a promising target for preventing and treating ovarian function decline related to reproductive aging.
Collapse
Affiliation(s)
- Ying Yao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Bin Wang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Reproductive Medicine and Embryo of Gansu, No. 1, Donggang West Road, Chengguan District, Lanzhou, Gansu Province, China
| | - Kaihua Yu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Ji Song
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Reproductive Medicine and Embryo of Gansu, No. 1, Donggang West Road, Chengguan District, Lanzhou, Gansu Province, China
| | - Liyan Wang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Reproductive Medicine and Embryo of Gansu, No. 1, Donggang West Road, Chengguan District, Lanzhou, Gansu Province, China
| | - Xuehong Zhang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China.
- Key Laboratory for Reproductive Medicine and Embryo of Gansu, No. 1, Donggang West Road, Chengguan District, Lanzhou, Gansu Province, China.
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, No. 1, Donggang West Road, Chengguan District, Lanzhou, Gansu Province, China.
| |
Collapse
|
46
|
Hense JD, Isola JVV, Garcia DN, Magalhães LS, Masternak MM, Stout MB, Schneider A. The role of cellular senescence in ovarian aging. NPJ AGING 2024; 10:35. [PMID: 39033161 PMCID: PMC11271274 DOI: 10.1038/s41514-024-00157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/29/2024] [Indexed: 07/23/2024]
Abstract
This review explores the relationship between ovarian aging and senescent cell accumulation, as well as the efficacy of senolytics to improve reproductive longevity. Reproductive longevity is determined by the age-associated decline in ovarian reserve, resulting in reduced fertility and eventually menopause. Cellular senescence is a state of permanent cell cycle arrest and resistance to apoptosis. Senescent cells accumulate in several tissues with advancing age, thereby promoting chronic inflammation and age-related diseases. Ovaries also appear to accumulate senescent cells with age, which might contribute to aging of the reproductive system and whole organism through SASP production. Importantly, senolytic drugs can eliminate senescent cells and may present a potential intervention to mitigate ovarian aging. Herein, we review the current literature related to the efficacy of senolytic drugs for extending the reproductive window in mice.
Collapse
Affiliation(s)
- Jéssica D Hense
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Driele N Garcia
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | | | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Augusto Schneider
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
47
|
Gu M, Wang Y, Yu Y. Ovarian fibrosis: molecular mechanisms and potential therapeutic targets. J Ovarian Res 2024; 17:139. [PMID: 38970048 PMCID: PMC11225137 DOI: 10.1186/s13048-024-01448-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 06/03/2024] [Indexed: 07/07/2024] Open
Abstract
Ovarian fibrosis, characterized by the excessive proliferation of ovarian fibroblasts and the accumulation of extracellular matrix (ECM), serves as one of the primary causes of ovarian dysfunction. Despite the critical role of ovarian fibrosis in maintaining the normal physiological function of the mammalian ovaries, research on this condition has been greatly underestimated, which leads to a lack of clinical treatment options for ovarian dysfunction caused by fibrosis. This review synthesizes recent research on the molecular mechanisms of ovarian fibrosis, encompassing TGF-β, extracellular matrix, inflammation, and other profibrotic factors contributing to abnormal ovarian fibrosis. Additionally, we summarize current treatment approaches for ovarian dysfunction targeting ovarian fibrosis, including antifibrotic drugs, stem cell transplantation, and exosomal therapies. The purpose of this review is to summarize the research progress on ovarian fibrosis and to propose potential therapeutic strategies targeting ovarian fibrosis for the treatment of ovarian dysfunction.
Collapse
Affiliation(s)
- Mengqing Gu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Yibo Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China.
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
- Institute of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China.
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
48
|
Xiao B, Dai Z, Li Z, Xu D, Yin H, Yang F, Sun N. Single-cell transcriptomic profiling unveils insights into ovarian fibrosis in obese mice. Biol Direct 2024; 19:52. [PMID: 38956667 PMCID: PMC11218254 DOI: 10.1186/s13062-024-00496-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Adiposity profoundly impacts reproductive health in both humans and animals. However, the precise subpopulations contributing to infertility under obese conditions remain elusive. RESULTS In this study, we established an obese mouse model through an eighteen-week high-fat diet regimen in adult female mice. Employing single-cell RNA sequencing (scRNA-seq), we constructed a comprehensive single-cell atlas of ovarian tissues from these mice to scrutinize the impact of obesity on the ovarian microenvironment. ScRNA-seq revealed notable alterations in the microenvironment of ovarian tissues in obese mice. Granulosa cells, stromal cells, T cells, and macrophages exhibited functional imbalances compared to the control group. We observed heightened interaction strength in the SPP1-CD44 pairing within lgfbp7+ granulosa cell subtypes and Il1bhigh monocyte subtypes in the ovarian tissues of obese mice. Moreover, the interaction strength between Il1bhigh monocyte subtypes and Pdgfrb+ stromal cell subtypes in the form of TNF - TNFrsf1α interaction was also enhanced subsequently to obesity, potentially contributing to ovarian fibrosis pathogenesis. CONCLUSIONS We propose a model wherein granulosa cells secrete SPP1 to activate monocytes, subsequently triggering TNF-α secretion by monocytes, thereby activating stromal cells and ultimately leading to the development of ovarian fibrosis. Intervening in this process may represent a promising avenue for improving clinical outcomes in fertility treatments for obese women.
Collapse
Affiliation(s)
- Bang Xiao
- Department of Medical Genetics, Naval Medical University, 800 Xiang yin Road, Shanghai, 200433, China
| | - Zhihui Dai
- Department of Medical Genetics, Naval Medical University, 800 Xiang yin Road, Shanghai, 200433, China
| | - Zhixuan Li
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, 51 Fu cheng Road, Beijing, 100853, China
| | - Dabing Xu
- Department of Reproductive Medicine, Second Affiliated Hospital of Naval Medical University, 415 Feng yang Road, Shanghai, 200003, China
| | - Haozan Yin
- Department of Medical Genetics, Naval Medical University, 800 Xiang yin Road, Shanghai, 200433, China
| | - Fu Yang
- Department of Medical Genetics, Naval Medical University, 800 Xiang yin Road, Shanghai, 200433, China.
- Department of Reproductive Medicine, Second Affiliated Hospital of Naval Medical University, 415 Feng yang Road, Shanghai, 200003, China.
| | - Ningxia Sun
- Department of Reproductive Medicine, Second Affiliated Hospital of Naval Medical University, 415 Feng yang Road, Shanghai, 200003, China.
| |
Collapse
|
49
|
Amargant F, Vieira C, Pritchard MT, Duncan FE. Systemic low-dose anti-fibrotic treatment attenuates ovarian aging in the mouse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600035. [PMID: 38979191 PMCID: PMC11230292 DOI: 10.1101/2024.06.21.600035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The female reproductive system is one of the first to age in humans, resulting in infertility and endocrine disruptions. The aging ovary assumes a fibro-inflammatory milieu which negatively impacts gamete quantity and quality as well as ovulation. Here we tested whether the systemic delivery of anti-inflammatory (Etanercept) or anti-fibrotic (Pirfenidone) drugs attenuates ovarian aging in mice. We first evaluated the ability of these drugs to decrease the expression of fibro-inflammatory genes in primary ovarian stromal cells. Whereas Etanercept did not block Tnf expression in ovarian stromal cells, Pirfenidone significantly reduced Col1a1 expression. We then tested Pirfenidone in vivo where the drug was delivered systemically via mini-osmotic pumps for 6-weeks. Pirfenidone mitigated the age-dependent increase in ovarian fibrosis without impacting overall health parameters. Ovarian function was improved in Pirfenidone-treated mice as evidenced by increased follicle and corpora lutea number, AMH levels, and improved estrous cyclicity. Transcriptomic analysis revealed that Pirfenidone treatment resulted in an upregulation of reproductive function-related genes at 8.5 months and a downregulation of inflammatory genes at 12 months of age. These findings demonstrate that reducing the fibroinflammatory ovarian microenvironment improves ovarian function, thereby supporting modulating the ovarian environment as a therapeutic avenue to extend reproductive longevity.
Collapse
Affiliation(s)
- Farners Amargant
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Carol Vieira
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michele T Pritchard
- Department of Pharmacology, Toxicology and Therapeutics and Institute for Reproductive and Developmental Sciences University of Kansas Medical Center, Kansas City, KS, USA
| | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
50
|
Zhang Z, Huang L, Brayboy L, Birrer M. Single-cell analysis of ovarian myeloid cells identifies aging associated changes in macrophages and signaling dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598667. [PMID: 38915572 PMCID: PMC11195259 DOI: 10.1101/2024.06.13.598667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The aging of mammalian ovary is accompanied by an increase in tissue fibrosis and heightened inflammation. Myeloid cells, including macrophages, monocytes, dendritic cells, and neutrophils, play pivotal roles in shaping the ovarian tissue microenvironment and regulating inflammatory responses. However, a comprehensive understanding of the roles of these cells in the ovarian aging process is lacking. To bridge this knowledge gap, we utilized single-cell RNA sequencing (scRNAseq) and flow cytometry analysis to functionally characterize CD45+ CD11b+ myeloid cell populations in young (3 months old) and aged (14-17 months old) murine ovaries. Our dataset unveiled the presence of five ovarian macrophage subsets, including a Cx3cr1 low Cd81 hi subset unique to the aged murine ovary. Most notably, our data revealed significant alterations in ANNEXIN and TGFβ signaling within aged ovarian myeloid cells, which suggest a novel mechanism contributing to the onset and progression of aging-associated inflammation and fibrosis in the ovarian tissue.
Collapse
Affiliation(s)
- Zijing Zhang
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR
- Winthrop P Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Lu Huang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Lynae Brayboy
- Department of Neuropediatrics Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Birrer
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR
- Winthrop P Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR
| |
Collapse
|