1
|
Gomes Paim LM, Bechstedt S. Regulation of microtubule growth rates and their impact on chromosomal instability. Cell Cycle 2025:1-20. [PMID: 40260826 DOI: 10.1080/15384101.2025.2485842] [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: 09/17/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 04/24/2025] Open
Abstract
Microtubules are polymers of α/β tubulin dimers that build the mitotic spindle, which segregates duplicated chromosomes during cell division. Microtubule function is governed by dynamic instability, whereby cycles of growth and shrinkage contribute to the forces necessary for chromosome movement. Regulation of microtubule growth velocity requires cell cycle-dependent changes in expression, localization and activity of microtubule-associated proteins (MAPs) as well as tubulin post-translational modifications that modulate microtubule dynamics. It has become clear that optimal microtubule growth velocities are required for proper chromosome segregation and ploidy maintenance. Suboptimal microtubule growth rates can result from altered activity of MAPs and could lead to aneuploidy, possibly by disrupting the establishment of microtubule bundles at kinetochores and altering the mechanical forces required for sister chromatid segregation. Future work using high-resolution, low-phototoxicity microscopy and novel fluorescent markers will be invaluable in obtaining deeper mechanistic insights into how microtubule processes contribute to chromosome segregation.
Collapse
Affiliation(s)
| | - Susanne Bechstedt
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
- Centre de Recherche en Biologie Structurale (CRBS), McGill University, Montréal, Canada
| |
Collapse
|
2
|
Castagnola L, Gallino L, Schafir A, Vota D, Grasso E, Gori S, Waschek J, Parborell F, Pérez Leirós C, Hauk V, Ramhorst R. Ovarian premature aging: VIP as key regulator of fibro-inflammation and foamy macrophages generation. Mol Cell Endocrinol 2025; 599:112486. [PMID: 39894337 DOI: 10.1016/j.mce.2025.112486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/21/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Ovarian aging is associated with fibro-inflammation, contributing to the decline in oocyte count and quality. Given the immunomodulatory properties of the vasoactive intestinal peptide (VIP) in the reproductive tract, we investigated its role in maintaining ovarian immune homeostasis and preventing premature aging. We evaluated young VIP knockout (KO) mice, comparing them to young wild type (WT) females, for signs of premature aging. Histological staining revealed aberrant ovarian morphology in VIP KO mice, characterized by increased atretic follicles and decreased ovarian reserve compared to WT controls. Moreover, VIP KO ovaries showed reduced vascularization, increased collagen deposition and elevated ROS and IL-1β levels. Foamy macrophages were significantly predominant, indicating premature aging in young VIP KO ovaries. To determine potential mechanisms behind these pathogenic changes, we conditioned peritoneal macrophages from young WT or VIP KO mice in vitro with ovarian-conditioned media from young WT or VIP KO mice to mimic the respective ovarian microenvironment. When WT or VIP KO peritoneal macrophages were conditioned with ovarian media from their respective genotypes, lipid droplet accumulation increased compared to control medium. In cross-genotype experiments, WT macrophages conditioned with media from VIP KO ovaries selectively accumulated higher levels of lipid droplets, whereas no differences were observed in VIP KO macrophages conditioned with WT ovarian media. This suggests that VIP KO macrophages are uniquely sensitized to the inflammatory environment of VIP KO ovaries, implicating both ovarian factors and macrophage status. These findings highlight the role of VIP in preventing fibro-inflammation, thereby preserving ovarian health and preventing premature aging.
Collapse
Affiliation(s)
- Lara Castagnola
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Lucila Gallino
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Ana Schafir
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Daiana Vota
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Esteban Grasso
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Soledad Gori
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - James Waschek
- The David Geffen School of Medicine, University of California, Los Angeles, USA
| | | | - Claudia Pérez Leirós
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Vanesa Hauk
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina.
| | - Rosanna Ramhorst
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina.
| |
Collapse
|
3
|
Gao M, Wang F, Xu T, Qiu Y, Cao T, Liu S, Wu W, Zhou Y, Liu H, Liu F, Huang J. Age-associated accumulation of RAB9 disrupts oocyte meiosis. Aging Cell 2025; 24:e14449. [PMID: 39676221 PMCID: PMC11984694 DOI: 10.1111/acel.14449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
The critical role of some RAB family members in oocyte meiosis has been extensively studied, but their role in oocyte aging remains poorly understood. Here, we report that the vesicle trafficking regulator, RAB9 GTPase, is essential for oocyte meiosis and aging in humans and mice. RAB9 was mainly located at the meiotic spindle periphery and cortex during oocyte meiosis. In humans and mice, we found that the RAB9 protein level were significantly increased in old oocytes. Age-related accumulation of RAB9 inhibits first polar body extrusion and reduces the developmental potential of oocytes. Further studies showed that increased Rab9 disrupts spindle formation and chromosome alignment. In addition, Rab9 overexpression disrupts the actin cap formation and reduces the cortical actin levels. Mechanically, Rab9-OE increases ROS levels, decreases mitochondrial membrane potential, ATP content and the mtDNA/nDNA ratio. Further studies showed that Rab9-OE activates the PINK1-PARKIN mitophagy pathway. Importantly, we found that reducing RAB9 protein expression in old oocytes could partially improve the rate of old oocyte maturation, ameliorate the accumulation of age-related ROS levels and spindle abnormalities, and partially rescue ATP levels, mtDNA/nDNA ratio, and PINK1 and PARKIN expression. In conclusion, our results suggest that RAB9 is required to maintain the balance between mitochondrial function and meiosis, and that reducing RAB9 expression is a potential strategy to ameliorate age-related deterioration of oocyte quality.
Collapse
Affiliation(s)
- Min Gao
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- Key Laboratory of Reproductive Health Diseases Research and Translation of Ministry of Education, The First Affiliated HospitalHainan Medical UniversityHaikouChina
| | - Fang Wang
- Department of Reproductive Medical CenterGuangdong Women and Children HospitalGuangzhouChina
| | - Tengteng Xu
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- Department of Gynecology, Clinical Transformation and Application Key Lab for Obstetrics and Gynecology, Pediatrics, and Reproductive Medicine of JiangmenJiangmen Central HospitalJiangmenChina
| | - Yanling Qiu
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
| | - Tianqi Cao
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
| | - Simiao Liu
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
| | - Wenlian Wu
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
| | - Yitong Zhou
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
| | - Haiying Liu
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
| | - Fenghua Liu
- Department of Reproductive Medical CenterGuangdong Women and Children HospitalGuangzhouChina
| | - Junjiu Huang
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital and School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
4
|
Zhou C, Zhang X, Xu G, Ran Y, Wang H, Xie X, Li A, Li F, Li X, Ding J, Zhang M, Sun Q, Ou X. A Microtubule-Associated Protein Functions in Preventing Oocytes from Evading the Spindle Assembly Checkpoint. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413097. [PMID: 39721007 PMCID: PMC11831433 DOI: 10.1002/advs.202413097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/02/2024] [Indexed: 12/28/2024]
Abstract
Aneuploidy eggs are a common cause of human infertility, spontaneous abortion, or trisomy syndromes. The spindle assembly checkpoint (SAC) plays a crucial role in preventing aneuploidy in oocytes, yet it is unclear if additional mechanisms exist to ensure oocyte adherence to this checkpoint. It is now revealed that the microtubule-associated protein NUSAP can prevent oocytes from evading the SAC and regulate the speed of the cell cycle. Mechanistically, the study identifies NUSAP as a novel stabilizer of the E3 ubiquitin ligase APC/CCDH1, protecting CDH1 from SCFBTRC-mediated degradation. Depletion of NUSAP reduces CDH1 protein level, leading to abnormal spindle assembly and chromosome alignment, and disrupting the balance of cell cycle proteins. This misregulated balance causes oocytes to evade the SAC. Consequently, these abnormal oocytes not only fail to arrest at metaphase but also accelerate the cell process, ultimately resulting in the production of aneuploid eggs. Together, the findings not only clarify the existence of mechanisms that ensure oocytes compliance with the spindle assembly checkpoint but also expand the new functions of NUSAP beyond its role as a microtubule- associated protein.
Collapse
Affiliation(s)
- Changyin Zhou
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
- School of Biomedical and Pharmaceutical SciencesGuangdong University of TechnologyGuangzhou510006China
| | - Xue Zhang
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
| | - Genlu Xu
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
- School of Biomedical and Pharmaceutical SciencesGuangdong University of TechnologyGuangzhou510006China
| | - Yuting Ran
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
- College of Animal Science and TechnologyAnhui Agricultural UniversityKey Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui ProvinceHefei230036China
| | - Hui Wang
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
- College of Animal Science and TechnologyAnhui Agricultural UniversityKey Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui ProvinceHefei230036China
| | - Xuefeng Xie
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
| | - Ang Li
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
| | - Fei Li
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
| | - Xiaozhen Li
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
| | - Jinlong Ding
- School of Biomedical and Pharmaceutical SciencesGuangdong University of TechnologyGuangzhou510006China
| | - Mianqun Zhang
- College of Animal Science and TechnologyAnhui Agricultural UniversityKey Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui ProvinceHefei230036China
| | - Qing‐Yuan Sun
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
| | - Xiang‐Hong Ou
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive HealthGuangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryReproductive Medicine CenterThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
| |
Collapse
|
5
|
Rong Y, Wu Y, Chen Y, Liu Q, Ai L, Wu Y, Zhu Y, Zhang Y, Liu C, Ma Y, Tong X, Jin J, Li X, Zhou Y, Ji S, Zhang S, Fan H. ZAR1/2-Regulated Epigenetic Modifications are Essential for Age-Associated Oocyte Quality Maintenance and Zygotic Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410305. [PMID: 39755931 PMCID: PMC11848533 DOI: 10.1002/advs.202410305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/23/2024] [Indexed: 01/06/2025]
Abstract
The developmental competence and epigenetic progression of oocytes gradually become dysregulated with increasing maternal age. However, the mechanisms underlying age-related epigenetic regulation in oocytes remain poorly understood. Zygote arrest proteins 1 and 2 (ZAR1/2) are two maternal factors with partially redundant roles in maintaining oocyte quality, mainly known by regulating mRNA stability. In addition to this known function, it is found that ZAR1/2 is required for oocyte epigenetic maturation and zygotic reprogramming. Zar1/2-deleted oocytes exhibited reduced levels of multiple histone modifications and of the expression of corresponding histone modifiers, along with over-condensed chromatin, leading to compromised minor zygotic genome activation and deficient embryo development following fertilization. Cytoplasmic ZAR1/2 participated in intranuclear epigenetic maturation by binding the transcripts encoding histone modifiers and regulating their stability and translational activity. Moreover, oocytes from aged mice exhibited similar histone-modification deficiencies as the Zar1/2-deleted oocytes. ZAR1/2 mRNA and protein levels are downregulated in oocytes from mice and women with advanced ages, suggesting ZAR1/2 as regulators of epigenetic changes with reproductive aging. This study presents a new nucleo-cytoplasmic interaction mechanism that is involved in preventing oocyte epigenetic aging. Further, ZAR1/2 represents potential gene targets for diagnosis and clinical interventions in age-associated deficiencies in oocyte and embryo development.
Collapse
Affiliation(s)
- Yan Rong
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Yu‐Ke Wu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Yingyan Chen
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Qing Liu
- Department of Traditional Chinese MedicineSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Leilei Ai
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Yun‐Wen Wu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Yezhang Zhu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Yin‐Li Zhang
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Chengkan Liu
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Yerong Ma
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Xiaomei Tong
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Jiamin Jin
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Xiaoxuan Li
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Yan Zhou
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Shu‐Yan Ji
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Songying Zhang
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Heng‐Yu Fan
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
- Center for Biomedical ResearchShaoxing InstituteZhejiang UniversityShaoxing312000China
| |
Collapse
|
6
|
Shapiro JG, Changela N, Jang JK, Joshi JN, McKim KS. Distinct checkpoint and homolog biorientation pathways regulate meiosis I in Drosophila oocytes. PLoS Genet 2025; 21:e1011400. [PMID: 39879252 PMCID: PMC11809923 DOI: 10.1371/journal.pgen.1011400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/10/2025] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
Mitosis and meiosis have two mechanisms for regulating the accuracy of chromosome segregation: error correction and the spindle assembly checkpoint (SAC). We have investigated the function of several checkpoint proteins in meiosis I of Drosophila oocytes. Increased localization of several SAC proteins was found upon depolymerization of microtubules by colchicine. However, unattached kinetochores or errors in biorientation of homologous chromosomes do not induce increased SAC protein localization. Furthermore, the metaphase I arrest does not depend on SAC genes, suggesting the APC is inhibited even if the SAC is not functional. Two SAC proteins, ROD of the ROD-ZW10-Zwilch (RZZ) complex and MPS1, are also required for the biorientation of homologous chromosomes during meiosis I, suggesting an error correction function. Both proteins aid in preventing or correcting erroneous attachments and depend on SPC105R for localization to the kinetochore. We have defined a region of SPC105R, amino acids 123-473, that is required for ROD localization and biorientation of homologous chromosomes at meiosis I. Surprisingly, ROD removal from kinetochores and movement towards spindle poles, termed "streaming," is independent of the dynein adaptor Spindly and is not linked to the stabilization of end-on attachments. Instead, meiotic RZZ streaming appears to depend on cell cycle stage and may be regulated independently of kinetochore attachment or biorientation status. We also show that Spindly is required for biorientation at meiosis I, and surprisingly, the direction of RZZ streaming.
Collapse
Affiliation(s)
- Joanatta G. Shapiro
- Waksman Institute, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Neha Changela
- Waksman Institute, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Janet K. Jang
- Waksman Institute, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Jay N. Joshi
- Waksman Institute, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Kim S. McKim
- Waksman Institute, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| |
Collapse
|
7
|
Kyogoku H. Chromosome segregation errors during early embryonic development. Reprod Med Biol 2025; 24:e12631. [PMID: 39845483 PMCID: PMC11751853 DOI: 10.1002/rmb2.12631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
Background Mitosis maintains a genome's genetic information in daughter cells by accurately segregating chromosomes. However, chromosome aberrations are common during early mammalian embryogenesis. Chromosomal abnormalities during the early stages of embryogenesis result in the formation of mosaic embryos, wherein cells with normal genomes coexist with cells exhibiting abnormal genomes. The precise frequency and etiology of such abnormalities remain unclear. It is postulated that these aberrations contribute to the etiology of a number of conditions, including infertility and congenital diseases such as Down's syndrome. Methods This review synthesizes current literature and data to elucidate the causes and implications of chromosome aberrations in early mammalian embryos. It places particular emphasis on identifying patterns of mosaicism and investigating the underlying mechanisms responsible for these abnormalities. Main Findings The underlying causes of chromosome abnormalities in early embryos were examined in the context of DNA replication and embryonic development. Conclusion A deeper understanding of chromosome abnormalities in early embryos could help develop new infertility treatments and advance research on cancers caused by these abnormalities. This article reviews current knowledge and gaps in understanding chromosome segregation abnormalities during embryogenesis and future directions in this field.
Collapse
|
8
|
Xiao W, Akao S, Otsuki J. Correlation between female pronuclear/cytoplasmic ratio and number of chromosomes in mouse zygotic stage: implications for aneuploidy assessment in ART. J Assist Reprod Genet 2025; 42:85-95. [PMID: 39585518 PMCID: PMC11805731 DOI: 10.1007/s10815-024-03312-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/03/2024] [Indexed: 11/26/2024] Open
Abstract
PURPOSE The objective of this study was to investigate the correlation between the pronuclear/cytoplasmic (PN/C) ratio and the number of chromosomes in mouse zygotes to understand the implications of pronuclear size regulation in early embryonic development. METHODS A combination of enucleation and aggregated chromosomes/chromatin (AC) transfer was utilized to create oocytes with varying numbers of chromosomes. Time-lapse imaging and immunofluorescence staining were employed to analyze pronuclear dynamics and chromosomal configurations. RESULTS Higher chromosome numbers correspond to a larger PN/C ratio. Oocytes with a higher number of chromosomes exhibited larger pronuclei. CONCLUSION The study underscores the complexity of pronuclear size regulation and its correlation with the number of chromosomes. The findings suggest potential applications in ART, where assessing the PN/C ratio could serve as a biomarker for zygote quality and aneuploidy.
Collapse
Affiliation(s)
- Wei Xiao
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, 1-1-1 Tsushimanaka, Kita, Okayama, 700-8530, Japan
| | - Sakura Akao
- Faculty of Agriculture, Okayama University, 1-1-1 Tsushimanaka, Kita, Okayama, 700-8530, Japan
| | - Junko Otsuki
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, 1-1-1 Tsushimanaka, Kita, Okayama, 700-8530, Japan.
- Assisted Reproductive Technology Center, Okayama University, 1-1-1 Tsushimanaka, Kita, Okayama, 700-8530, Japan.
| |
Collapse
|
9
|
Fan LH, Qi ST, Wang ZB, Ouyang YC, Lei WL, Wang Y, Li A, Wang F, Li J, Li L, Li YY, Hou Y, Schatten H, Wang WH, Sun QY, Ou XH. MEIKIN expression and its C-terminal phosphorylation by PLK1 is closely related the metaphase-anaphase transition by affecting cyclin B1 and Securin stabilization in meiotic oocyte. Histochem Cell Biol 2024; 162:447-464. [PMID: 39093409 DOI: 10.1007/s00418-024-02316-7] [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] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Oocyte meiotic maturation failure and chromosome abnormality is one of the main causes of infertility, abortion, and diseases. The mono-orientation of sister chromatids during the first meiosis is important for ensuring accurate chromosome segregation in oocytes. MEIKIN is a germ cell-specific protein that can regulate the mono-orientation of sister chromatids and the protection of the centromeric cohesin complex during meiosis I. Here we found that MEIKIN is a maternal protein that was highly expressed in mouse oocytes before the metaphase I (MI) stage, but became degraded by the MII stage and dramatically reduced after fertilization. Strikingly, MEIKIN underwent phosphorylation modification after germinal vesicle breakdown (GVBD), indicating its possible function in subsequent cellular event regulation. We further showed that MEIKIN phosphorylation was mediated by PLK1 at its carboxyl terminal region and its C-terminus was its key functional domain. To clarify the biological significance of meikin degradation during later stages of oocyte maturation, exogenous expression of MEIKIN was employed, which showed that suppression of MEIKIN degradation resulted in chromosome misalignment, cyclin B1 and Securin degradation failure, and MI arrest through a spindle assembly checkpoint (SAC)-independent mechanism. Exogenous expression of MEIKIN also inhibited metaphase II (MII) exit and early embryo development. These results indicate that proper MEIKIN expression level and its C-terminal phosphorylation by PLK1 are critical for regulating the metaphase-anaphase transition in meiotic oocyte. The findings of this study are important for understanding the regulation of chromosome segregation and the prevention meiotic abnormality.
Collapse
Affiliation(s)
- Li-Hua Fan
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shu-Tao Qi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Major Obstetrics Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wen-Long Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yue Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ang Li
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Feng Wang
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Jian Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Li Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuan-Yuan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Wei-Hua Wang
- Key Laboratory of Major Obstetrics Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Qing-Yuan Sun
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
- University of Chinese Academy of Sciences, Beijing, 100101, China.
| | - Xiang-Hong Ou
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
| |
Collapse
|
10
|
Biswas L, Tyc KM, Aboelenain M, Sun S, Dundović I, Vukušić K, Liu J, Guo V, Xu M, Scott RT, Tao X, Tolić IM, Xing J, Schindler K. Maternal genetic variants in kinesin motor domains prematurely increase egg aneuploidy. Proc Natl Acad Sci U S A 2024; 121:e2414963121. [PMID: 39475646 PMCID: PMC11551467 DOI: 10.1073/pnas.2414963121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/27/2024] [Indexed: 11/06/2024] Open
Abstract
The female reproductive lifespan is highly dependent on egg quality, especially the presence of a normal number of chromosomes in an egg, known as euploidy. Mistakes in meiosis leading to egg aneuploidy are frequent in humans. Yet, knowledge of the precise genetic landscape that causes egg aneuploidy in women is limited, as phenotypic data on the frequency of human egg aneuploidy are difficult to obtain and therefore absent in public genetic datasets. Here, we identify genetic determinants of reproductive aging via egg aneuploidy in women using a biobank of individual maternal exomes linked with maternal age and embryonic aneuploidy data. Using the exome data, we identified 404 genes bearing variants enriched in individuals with pathologically elevated egg aneuploidy rates. Analysis of the gene ontology and protein-protein interaction network implicated genes encoding the kinesin protein family in egg aneuploidy. We interrogate the causal relationship of the human variants within candidate kinesin genes via experimental perturbations and demonstrate that motor domain variants increase aneuploidy in mouse oocytes. Finally, using a knock-in mouse model, we validate that a specific variant in kinesin KIF18A accelerates reproductive aging and diminishes fertility. These findings reveal additional functional mechanisms of reproductive aging and shed light on how genetic variation underlies individual heterogeneity in the female reproductive lifespan, which might be leveraged to predict reproductive longevity. Together, these results lay the groundwork for the noninvasive biomarkers for egg quality, a first step toward personalized fertility medicine.
Collapse
Affiliation(s)
- Leelabati Biswas
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Katarzyna M. Tyc
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Mansour Aboelenain
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Department of Theriogenology, Faculty of Veterinary Medicine, Mansoura University, Mansoura35516, Egypt
| | - Siqi Sun
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Iva Dundović
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb1000, Croatia
| | - Kruno Vukušić
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb1000, Croatia
| | - Jason Liu
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | | | - Min Xu
- Department of Statistics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | | | - Xin Tao
- Juno Genetics US, Basking Ridge, NJ07920
| | - Iva M. Tolić
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb1000, Croatia
| | - Jinchuan Xing
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| |
Collapse
|
11
|
Wu T, Luo Y, Zhang M, Chen B, Du X, Gu H, Xie S, Pan Z, Yu R, Hai R, Niu X, Hao G, Jin L, Shi J, Sun X, Kuang Y, Li W, Sang Q, Wang L. Mechanisms of minor pole-mediated spindle bipolarization in human oocytes. Science 2024; 385:eado1022. [PMID: 39172836 DOI: 10.1126/science.ado1022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/29/2024] [Accepted: 07/02/2024] [Indexed: 08/24/2024]
Abstract
Spindle bipolarization, the process of a microtubule mass transforming into a bipolar spindle, is a prerequisite for accurate chromosome segregation. In contrast to mitotic cells, the process and mechanism of spindle bipolarization in human oocytes remains unclear. Using high-resolution imaging in more than 1800 human oocytes, we revealed a typical state of multipolar intermediates that form during spindle bipolarization and elucidated the mechanism underlying this process. We found that the minor poles formed in multiple kinetochore clusters contribute to the generation of multipolar intermediates. We further determined the essential roles of HAUS6, KIF11, and KIF18A in spindle bipolarization and identified mutations in these genes in infertile patients characterized by oocyte or embryo defects. These results provide insights into the physiological and pathological mechanisms of spindle bipolarization in human oocytes.
Collapse
Affiliation(s)
- Tianyu Wu
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Yuxi Luo
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Meiling Zhang
- Center for Reproductive Medicine and Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Fudan University, Shanghai 200032, China
| | - Xingzhu Du
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Hao Gu
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Siyuan Xie
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Zhiqi Pan
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Ran Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Ruiqi Hai
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Xiangli Niu
- Reproductive Hospital of Guangxi Zhuang Autonomous Region, Nanning 530029, China
| | - Guimin Hao
- Hebei Clinical Research Center for Birth Defects, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Liping Jin
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Juanzi Shi
- Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an 710003, China
| | - Xiaoxi Sun
- Shanghai JIAI Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Wen Li
- Center for Reproductive Medicine and Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| |
Collapse
|
12
|
Shapiro JG, Changela N, Jang JK, Joshi JN, McKim KS. Distinct checkpoint and homolog biorientation pathways regulate meiosis I in Drosophila oocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608908. [PMID: 39229242 PMCID: PMC11370425 DOI: 10.1101/2024.08.21.608908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Mitosis and meiosis have two mechanisms for regulating the accuracy of chromosome segregation: error correction and the spindle assembly checkpoint (SAC). We have investigated the function of several checkpoint proteins in meiosis I of Drosophila oocytes. Evidence of a SAC response by several of these proteins is found upon depolymerization of microtubules by colchicine. However, unattached kinetochores or errors in biorientation of homologous chromosomes does not induce a SAC response. Furthermore, the metaphase I arrest does not depend on SAC genes, suggesting the APC is inhibited even if the SAC is silenced. Two SAC proteins, ROD of the ROD-ZW10-Zwilch (RZZ) complex and MPS1, are also required for the biorientation of homologous chromosomes during meiosis I, suggesting an error correction function. Both proteins aid in preventing or correcting erroneous attachments and depend on SPC105R for localization to the kinetochore. We have defined a region of SPC105R, amino acids 123-473, that is required for ROD localization and biorientation of homologous chromosomes at meiosis I. Surprisingly, ROD removal, or "streaming", is independent of the dynein adaptor Spindly and is not linked to the stabilization of end-on attachments. Instead, meiotic RZZ streaming appears to depend on cell cycle stage and may be regulated independently of kinetochore attachment or biorientation status. We also show that dynein adaptor Spindly is also required for biorientation at meiosis I, and surprisingly, the direction of RZZ streaming.
Collapse
Affiliation(s)
- Joanatta G Shapiro
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Neha Changela
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Janet K Jang
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Jay N Joshi
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Kim S McKim
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, New Jersey, United States of America
| |
Collapse
|
13
|
Huang W, Li X, Yang H, Huang H. The impact of maternal age on aneuploidy in oocytes: Reproductive consequences, molecular mechanisms, and future directions. Ageing Res Rev 2024; 97:102292. [PMID: 38582380 DOI: 10.1016/j.arr.2024.102292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/12/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Age-related aneuploidy in human oocytes is a major factor contributing to decreased fertility and adverse reproductive outcomes. As females age, their oocytes are more prone to meiotic chromosome segregation errors, leading primarily to aneuploidy. Elevated aneuploidy rates have also been observed in oocytes from very young, prepubertal conceptions. A key barrier to developing effective treatments for age-related oocyte aneuploidy is our incomplete understanding of the molecular mechanisms involved. The challenge is becoming increasingly critical as more people choose to delay childbearing, a trend that has significant societal implications. In this review, we summarize current knowledge regarding the process of oocyte meiosis and folliculogenesis, highlighting the relationship between age and chromosomal aberrations in oocytes and embryos, and integrate proposed mechanisms of age-related meiotic disturbances across structural, protein, and genomic levels. Our goal is to spur new research directions and therapeutic avenues.
Collapse
Affiliation(s)
- Weiwei Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Xinyuan Li
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Hongbo Yang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Department of Obstetrics and Gynecology, International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
14
|
Butkiewicz AF, Amaral A, Cerveira-Pinto M, Kordowitzki P. Assessing the Influence of Maternal Age in Bovine Embryos and Oocytes: A Model for Human Reproductive Aging. Aging Dis 2024; 16:757-768. [PMID: 38916737 PMCID: PMC11964423 DOI: 10.14336/ad.2024.0305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/05/2024] [Indexed: 06/26/2024] Open
Abstract
In the first weeks after fertilization, embryo mortality in cattle is significantly higher. It is well known that the age of the dam is one of the crucial factors affecting the quality of embryos and oocytes in many mammalian species. In older cattle, there are several evidences that embryo quality decreases, due to a decrease in ovarian reserve, a decrease in mtDNA and ATP, a decrease in progesterone levels, and due to susceptibility to genetic mutations. Herein, we intend to provide an updated summary of recent research on the effects of maternal age on embryos and oocytes of domestic cattle which are a widely used model species for human oocytes and early embryonic development.
Collapse
Affiliation(s)
- Aleksander Franciszek Butkiewicz
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland.
| | - Ana Amaral
- CIISA—Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal.
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal.
| | - Marta Cerveira-Pinto
- CIISA—Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal.
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal.
| | - Pawel Kordowitzki
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland.
- Department of Gynaecology, European Competence Center for Ovarian Cancer, Charité, Berlin, Germany.
| |
Collapse
|
15
|
Goud PT, Goud AP, Camp OG, Bai D, Gonik B, Diamond MP, Abu-Soud HM. Chronological age enhances aging phenomena and protein nitration in oocyte. Front Endocrinol (Lausanne) 2023; 14:1251102. [PMID: 38149097 PMCID: PMC10749940 DOI: 10.3389/fendo.2023.1251102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Background The average age of childbearing has increased over the years contributing to infertility, miscarriages, and chromosomal abnormalities largely invoked by an age-related decline in oocyte quality. In this study, we investigate the role of nitric oxide (NO) insufficiency and protein nitration in oocyte chronological aging. Methods Mouse oocytes were retrieved from young breeders (YB, 8-14 weeks [w]), retired breeders (RB, 48-52w) and old animals (OA, 80-84w) at 13.5 and 17 hours after ovulation trigger. They were assessed for zona pellucida dissolution time (ZPDT); ooplasmic microtubule dynamics (OMD); cortical granule (CG) status and spindle morphology (SM), as markers of oocyte quality. Sibling oocytes from RB were exposed to NO supplementation and assessed for aging phenomena (AP). All oocyte cumulus complexes were subjected to fluorescence nitrotyrosine (NT) immunocytochemistry and confocal microscopy to assess morphology and protein nitration. Results At 13.5 h from hCG trigger, oocytes from RB compared to YB had significantly increased ZPDT (37.8 ± 11.9 vs 22.1 ± 4.1 seconds [s]), OMD (46.9 vs 0%), CG loss (39.4 vs 0%), and decreased normal SM (30.3 vs 81.3%), indicating premature AP that worsened among oocytes from RB at 17 hours post-hCG trigger. When exposed to SNAP, RB AP significantly decreased (ZPDT: 35.1 ± 5.5 vs 46.3 ± 8.9s, OMD: 13.3 vs 75.0% and CG loss: 50.0 vs 93.3%) and SM improved (80.0 vs 14.3%). The incidence of NT positivity was significantly higher in cumulus cells (13.5 h, 46.7 ± 4.5 vs 3.4 ± 0.7%; 17 h, 82.2 ± 2.9 vs 23.3 ± 3.6%) and oocytes (13.5 h, 57.1 vs 0%; 17 h, 100.0 vs 55.5%) from RB compared to YB. Oocytes retrieved decreased with advancing age (29.8 ± 4.1 per animal in the YB group compared to 10.2 ± 2.1 in RB and 4.0 ± 1.6 in OA). Oocytes from OA displayed increased ZPDT, major CG loss, increased OMD and spindle abnormalities, as well as pronuclear formation, confirming spontaneous meiosis to interphase transition. Conclusions Oocytes undergo zona pellucida hardening, altered spindle and ooplasmic microtubules, and premature cortical granule release, indicative of spontaneous meiosis-interphase transition, as a function of chronological aging. These changes are also associated with NO insufficiency and protein nitration and may be alleviated through supplementation with an NO-donor.
Collapse
Affiliation(s)
- Pravin T. Goud
- Laurel Fertility Center, San Francisco, CA, United States
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of California Davis Medical School, Sacramento, CA, United States
- Department of Obstetrics and Gynecology, University of California Davis Medical School, Sacramento, CA, United States
| | - Anuradha P. Goud
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States
| | - Olivia G. Camp
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States
| | - David Bai
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States
| | - Bernard Gonik
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Michael P. Diamond
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Augusta University, Augusta, GA, United States
| | - Husam M. Abu-Soud
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
16
|
Dong J, Jin L, Bao S, Chen B, Zeng Y, Luo Y, Du X, Sang Q, Wu T, Wang L. Ectopic expression of human TUBB8 leads to increased aneuploidy in mouse oocytes. Cell Discov 2023; 9:105. [PMID: 37875488 PMCID: PMC10598138 DOI: 10.1038/s41421-023-00599-z] [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: 02/07/2023] [Accepted: 08/26/2023] [Indexed: 10/26/2023] Open
Abstract
Aneuploidy seriously compromises female fertility and increases incidence of birth defects. Rates of aneuploidy in human eggs from even young women are significantly higher than those in other mammals. However, intrinsic genetic factors underlying this high incidence of aneuploidy in human eggs remain largely unknown. Here, we found that ectopic expression of human TUBB8 in mouse oocytes increases rates of aneuploidy by causing kinetochore-microtubule (K-MT) attachment defects. Stretched bivalents in mouse oocytes expressing TUBB8 are under less tension, resulting in continuous phosphorylation status of HEC1 by AURKB/C at late metaphase I that impairs the established correct K-MT attachments. This reduced tension in stretched bivalents likely correlates with decreased recruitment of KIF11 on meiotic spindles. We also found that ectopic expression of TUBB8 without its C-terminal tail decreases aneuploidy rates by reducing erroneous K-MT attachments. Importantly, variants in the C-terminal tail of TUBB8 were identified in patients with recurrent miscarriages. Ectopic expression of an identified TUBB8 variant in mouse oocytes also compromises K-MT attachments and increases aneuploidy rates. In conclusion, our study provides novel understanding for physiological mechanisms of aneuploidy in human eggs as well as for pathophysiological mechanisms involved in recurrent miscarriages.
Collapse
Affiliation(s)
- Jie Dong
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Liping Jin
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shihua Bao
- Department of Reproductive Immunology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Fudan University, Shanghai, China
| | - Yang Zeng
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Yuxi Luo
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Xingzhu Du
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| | - Tianyu Wu
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Muhammad T, Wan Y, Lv Y, Li H, Naushad W, Chan WY, Lu G, Chen ZJ, Liu H. Maternal obesity: A potential disruptor of female fertility and current interventions to reduce associated risks. Obes Rev 2023; 24:e13603. [PMID: 37452501 DOI: 10.1111/obr.13603] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 05/17/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
Currently, obesity has achieved epidemic levels in reproductive-aged women with a myriad of consequences. Obesity is susceptible to several reproductive complications that eventually affect fertility rates. These complications originate from the deteriorated quality of oocytes from mothers with obesity, which increases the probability of chromosomal aneuploidy, elevated reactive oxygen species production, compromised embryonic developmental competency, and eventually reduced fertility. Maternal obesity is linked to pregnancy complications such as implantation error, abortion, miscarriage, and early pregnancy loss. This review highlights the adverse effects of maternal obesity on female fertility, with a focus on the mechanistic link between maternal obesity and oocyte quality and discusses possible measures to reduce its associated risks.
Collapse
Affiliation(s)
- Tahir Muhammad
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Pakistan
| | - Yanling Wan
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
| | - Yue Lv
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Hanzhen Li
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
| | - Wasifa Naushad
- Department of Pathology, Microbiology and Immunology, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA
| | - Wai-Yee Chan
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Gang Lu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, 999077, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200000, China
| | - Hongbin Liu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, 999077, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, China
| |
Collapse
|
18
|
Ferreira AF, Soares M, Almeida-Santos T, Ramalho-Santos J, Sousa AP. Aging and oocyte competence: A molecular cell perspective. WIREs Mech Dis 2023; 15:e1613. [PMID: 37248206 DOI: 10.1002/wsbm.1613] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 12/30/2022] [Accepted: 04/19/2023] [Indexed: 05/31/2023]
Abstract
Follicular microenvironment is paramount in the acquisition of oocyte competence, which is dependent on two interconnected and interdependent processes: nuclear and cytoplasmic maturation. Extensive research conducted in human and model systems has provided evidence that those processes are disturbed with female aging. In fact, advanced maternal age (AMA) is associated with a lower chance of pregnancy and live birth, explained by the age-related decline in oocyte quality/competence. This decline has largely been attributed to mitochondria, essential for oocyte maturation, fertilization, and embryo development; with mitochondrial dysfunction leading to oxidative stress, responsible for nuclear and mitochondrial damage, suboptimal intracellular energy levels, calcium disturbance, and meiotic spindle alterations, that may result in oocyte aneuploidy. Nuclear-related mechanisms that justify increased oocyte aneuploidy include deoxyribonucleic acid (DNA) damage, loss of chromosomal cohesion, spindle assembly checkpoint dysfunction, meiotic recombination errors, and telomere attrition. On the other hand, age-dependent cytoplasmic maturation failure is related to mitochondrial dysfunction, altered mitochondrial biogenesis, altered mitochondrial morphology, distribution, activity, and dynamics, dysmorphic smooth endoplasmic reticulum and calcium disturbance, and alterations in the cytoskeleton. Furthermore, reproductive somatic cells also experience the effects of aging, including mitochondrial dysfunction and DNA damage, compromising the crosstalk between granulosa/cumulus cells and oocytes, also affected by a loss of gap junctions. Old oocytes seem therefore to mature in an altered microenvironment, with changes in metabolites, ribonucleic acid (RNA), proteins, and lipids. Overall, understanding the mechanisms implicated in the loss of oocyte quality will allow the establishment of emerging biomarkers and potential therapeutic anti-aging strategies. This article is categorized under: Reproductive System Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Ana Filipa Ferreira
- Reproductive Medicine Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculty of Medicine, Azinhaga de Santa Comba, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, CIBB, University of Coimbra, Coimbra, Portugal
| | - Maria Soares
- CNC-Center for Neuroscience and Cell Biology, CIBB, University of Coimbra, Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Teresa Almeida-Santos
- Reproductive Medicine Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculty of Medicine, Azinhaga de Santa Comba, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, CIBB, University of Coimbra, Coimbra, Portugal
| | - João Ramalho-Santos
- CNC-Center for Neuroscience and Cell Biology, CIBB, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, Calçada Martim de Freitas, University of Coimbra, Coimbra, Portugal
| | - Ana Paula Sousa
- Reproductive Medicine Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, CIBB, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
19
|
Ruan JL, Liang SS, Pan JP, Chen ZQ, Teng XM. Artificial oocyte activation with Ca 2+ ionophore improves reproductive outcomes in patients with fertilization failure and poor embryo development in previous ICSI cycles. Front Endocrinol (Lausanne) 2023; 14:1244507. [PMID: 37635975 PMCID: PMC10455927 DOI: 10.3389/fendo.2023.1244507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Research question Does artificial oocyte activation (AOA) by a calcium ionophore (ionomycin) improve the previous fertilization failure or poor embryo development of intracytoplasmic sperm injection (ICSI) account for male factor infertility or other infertility causes? Design This retrospective study involved 114 patients receiving ICSI-AOA in Shanghai First Maternity and Infant Hospital with previous ICSI fertilization failure or poor embryo development. The previous ICSI cycles of the same patients without AOA served as the control group. The fertilization rates, cleavage rates, transferable embryo rates and blastocyst formation rates of the two groups were compared. Additionally, the clinical pregnancy, implantation rate and live birth rates were also compared to assess the efficiency and safety of AOA. Furthermore, two subgroup analyses were performed in this study based on the cause of infertility and the reason for AOA. The fertilization rate, embryonic development potential and clinical outcome were compared among groups. Results Among 114 ICSI-AOA cycles, the fertilization rate, top-quality embryo rate, implantation rate, clinical pregnancy per patient and live birth rate per patient were improved significantly compared with previous ICSI cycles (p<0.05 to P< 0.001), and the miscarriage rate in the AOA group was significantly lower than that of the control group (p<0.001). In the AOA subgroups based on the cause of infertility, the fertilization rates of each subgroup were significantly improved compared with previous control cycles except for the mixed factor infertility subgroup (p<0.05 to p<0.001). In the AOA subgroups based on the reason for AOA, the fertilization rates of each subgroup were significantly increased compared with those in their previous ICSI cycle without AOA (p<0.001); however, there was no significant difference in the top-quality embryo rate. No significant improvement was found in the implantation rates and the clinical pregnancy rate in each subgroup except for the poor embryo development subgroup. In the 114 AOA cycles, 35 healthy infants (21 singletons and 7 twins) were delivered without major congenital birth defects or malformations. Conclusion This study showed that AOA with the calcium ionophore ionomycin can improve the reproductive outcomes of patients with previous fertilization failure and poor embryo development after ICSI.
Collapse
Affiliation(s)
- Jing Ling Ruan
- Reproductive Medicine Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shan Shan Liang
- Reproductive Medicine Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jia Ping Pan
- Reproductive Medicine Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhi Qin Chen
- Reproductive Medicine Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiao Ming Teng
- Reproductive Medicine Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
20
|
Qiao JY, Zhou Q, Xu K, Yue W, Lei WL, Li YY, Gu LJ, Ouyang YC, Hou Y, Schatten H, Meng TG, Wang ZB, Sun QY. Mad2 is dispensable for accurate chromosome segregation but becomes essential when oocytes are subjected to environmental stress. Development 2023; 150:dev201398. [PMID: 37485540 DOI: 10.1242/dev.201398] [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: 10/27/2022] [Accepted: 06/20/2023] [Indexed: 07/25/2023]
Abstract
Accurate chromosome segregation, monitored by the spindle assembly checkpoint (SAC), is crucial for the production of euploid cells. Previous in vitro studies by us and others showed that Mad2, a core member of the SAC, performs a checkpoint function in oocyte meiosis. Here, through an oocyte-specific knockout approach in mouse, we reconfirmed that Mad2-deficient oocytes exhibit an accelerated metaphase-to-anaphase transition caused by premature degradation of securin and cyclin B1 and subsequent activation of separase in meiosis I. However, it was surprising that the knockout mice were completely fertile and the resulting oocytes were euploid. In the absence of Mad2, other SAC proteins, including BubR1, Bub3 and Mad1, were normally recruited to the kinetochores, which likely explains the balanced chromosome separation. Further studies showed that the chromosome separation in Mad2-null oocytes was particularly sensitive to environmental changes and, when matured in vitro, showed chromosome misalignment, lagging chromosomes, and aneuploidy with premature separation of sister chromatids, which was exacerbated at a lower temperature. We reveal for the first time that Mad2 is dispensable for proper chromosome segregation but acts to mitigate environmental stress in meiotic oocytes.
Collapse
Affiliation(s)
- Jing-Yi Qiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhou
- Department of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Ke Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Yue
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wen-Long Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuan-Yuan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lin-Jian Gu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Tie-Gang Meng
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qing-Yuan Sun
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China
| |
Collapse
|
21
|
Gu Y, Xu J, Sun F, Cheng J. Elevated intracellular pH of zygotes during mouse aging causes mitochondrial dysfunction associated with poor embryo development. Mol Cell Endocrinol 2023:111991. [PMID: 37336488 DOI: 10.1016/j.mce.2023.111991] [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: 02/08/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023]
Abstract
The mortality of preimplantation embryos is positively correlated with maternal age. However, the underlying mechanism for the poor quality of embryos remains unclear. Here, we found that aging caused elevated intracellular pH (pHi) in zygotes, which could trigger aberrant mitochondrial membrane potential, increased reactive oxygen species (ROS) levels, and poor embryo development. Moreover, single-cell transcriptome sequencing of mouse zygotes identified 120 genes that were significantly differentially expressed (DE) between young and older zygotes. These include genes such as Slc14a1, Fxyd5, CD74, and Bst, which are related to cell division, ion transporter, and cell differentiation. Further analysis indicated that these DE genes were enriched in apoptosis, the NF-kappa B signaling pathway, and the chemokine signaling pathway, which might be the key regulatory pathway affecting the quality of zygotes and subsequent embryo development. Taken together, our study helps elucidate the poor quality and development of older preimplantation embryos.
Collapse
Affiliation(s)
- Yimin Gu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Junjie Xu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China; Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, 7, Taiyuan, 030001, China
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Jinmei Cheng
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| |
Collapse
|
22
|
Abdulsamad HMR, Murtaza ZF, AlMuhairi HM, Bafleh WS, AlMansoori SA, AlQubaisi SA, Hamdan H, Kashir J. The Therapeutic and Diagnostic Potential of Phospholipase C Zeta, Oocyte Activation, and Calcium in Treating Human Infertility. Pharmaceuticals (Basel) 2023; 16:441. [PMID: 36986540 PMCID: PMC10056371 DOI: 10.3390/ph16030441] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/19/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Oocyte activation, a fundamental event during mammalian fertilisation, is initiated by concerted intracellular patterns of calcium (Ca2+) release, termed Ca2+ oscillations, predominantly driven by testis-specific phospholipase C zeta (PLCζ). Ca2+ exerts a pivotal role in not just regulating oocyte activation and driving fertilisation, but also in influencing the quality of embryogenesis. In humans, a failure of Ca2+ release, or defects in related mechanisms, have been reported to result in infertility. Furthermore, mutations in the PLCζ gene and abnormalities in sperm PLCζ protein and RNA, have been strongly associated with forms of male infertility where oocyte activation is deficient. Concurrently, specific patterns and profiles of PLCζ in human sperm have been linked to parameters of semen quality, suggesting the potential for PLCζ as a powerful target for both therapeutics and diagnostics of human fertility. However, further to PLCζ and given the strong role played by Ca2+ in fertilisation, targets down- and up-stream of this process may also present a significantly similar level of promise. Herein, we systematically summarise recent advancements and controversies in the field to update expanding clinical associations between Ca2+-release, PLCζ, oocyte activation and human fertility. We discuss how such associations may potentially underlie defective embryogenesis and recurrent implantation failure following fertility treatments, alongside potential diagnostic and therapeutic avenues presented by oocyte activation for the diagnosis and treatment of human infertility.
Collapse
Affiliation(s)
- Haia M. R. Abdulsamad
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Zoha F. Murtaza
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Hessa M. AlMuhairi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Wjdan S. Bafleh
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Salma A. AlMansoori
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Shaikha A. AlQubaisi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Hamdan Hamdan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Junaid Kashir
- Department of Biology, College of Arts and Science, Khalifa University, Abu Dhabi 127788, United Arab Emirates
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia
| |
Collapse
|
23
|
KYOGOKU H, KITAJIMA TS. The large cytoplasmic volume of oocyte. J Reprod Dev 2023; 69:1-9. [PMID: 36436912 PMCID: PMC9939283 DOI: 10.1262/jrd.2022-101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The study of the size of cells and organelles has a long history, dating back to the 1600s when cells were defined. In particular, various methods have elucidated the size of the nucleus and the mitotic spindle in several species. However, little research has been conducted on oocyte size and organelles in mammals, and many questions remain to be answered. The appropriate size is essential to cell function properly. Oocytes have a very large cytoplasm, which is more than 100 times larger than that of general somatic cells in mammals. In this review, we discuss how oocytes acquire an enormous cytoplasmic size and the adverse effects of a large cytoplasmic size on cellular functions.
Collapse
Affiliation(s)
- Hirohisa KYOGOKU
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan,Laboratory for Chromosome Segregation, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Tomoya S KITAJIMA
- Laboratory for Chromosome Segregation, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| |
Collapse
|
24
|
Gallino L, Hauk V, Castagnola L, Vota D, Pascuali N, Parborell F, May M, Fontana V, Merech F, Naguila Z, Waschek J, Leirós CP, Ramhorst R. Vasoactive intestinal peptide deficiency promotes ovarian dysfunction associated to a proinflammatory microenvironment reminiscent of premature aging. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166585. [PMID: 36423894 DOI: 10.1016/j.bbadis.2022.166585] [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: 03/07/2022] [Revised: 10/15/2022] [Accepted: 10/20/2022] [Indexed: 11/23/2022]
Abstract
Complex immune regulation during pregnancy is required to ensure a successful pregnancy outcome. Vasoactive intestinal peptide (VIP) has local immunoregulatory effects on the ovary, uterus and maternal-fetal interface that favor a tolerogenic maternal microenvironment. Since the VIP Knockout (KO) mice are subfertile, we investigated the mechanisms underlying the effects of VIP deficiency on ovarian physiology and immune homeostasis. Therefore, we studied VIP KO, deficient (HT) and wild type (WT) female mice in estrus at 3 or 8 months of age. Young KO mice showed abnormal cycle timing and regularity associated with dysfunctional ovaries. Ovaries presented higher number of atretic follicles and reduced number of corpora lutea leading to a lower ovulation rates. Part of the VIP KO mice (25 %) failed to ovulate or ovulated oocytes incompetent to be fertilized (50 %). In particular, ovaries of young KO mice exhibited features of premature aging accompanied by a pro-inflammatory milieu with increased levels of IL-1β. A unique macrophage subpopulation identified as "foamy macrophages" was found. On the other hand, aged VIP KO females did not gain body weight probably due to the sustained production of E2. Finally, the adoptive transfer of FOXP3+ cells to infertile VIP KO females resulted in their selective recruitment to the ovary. It increased FOXP3/RORγt and TGFβ/IL-6 ratio improving ovarian microenvironment and pregnancy rate. The present results suggest that VIP contributes to ovarian homeostatic mechanisms required for a successful pregnancy.
Collapse
Affiliation(s)
- Lucila Gallino
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Vanesa Hauk
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Lara Castagnola
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Daiana Vota
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | | | | | - Maria May
- ININFA, Universidad de Buenos Aires, CONICET, Argentina
| | - Vanina Fontana
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Fatima Merech
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Zaira Naguila
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - James Waschek
- The David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Claudia Perez Leirós
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Rosanna Ramhorst
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina.
| |
Collapse
|
25
|
Takahashi N, Franciosi F, Daldello EM, Luong XG, Althoff P, Wang X, Conti M. CPEB1-dependent disruption of the mRNA translation program in oocytes during maternal aging. Nat Commun 2023; 14:416. [PMID: 36697412 PMCID: PMC9877008 DOI: 10.1038/s41467-023-35994-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/11/2023] [Indexed: 01/27/2023] Open
Abstract
The molecular causes of deteriorating oocyte quality during aging are poorly defined. Since oocyte developmental competence relies on post-transcriptional regulations, we tested whether defective mRNA translation contributes to this decline in quality. Disruption in ribosome loading on maternal transcripts is present in old oocytes. Using a candidate approach, we detect altered translation of 3'-UTR-reporters and altered poly(A) length of the endogenous mRNAs. mRNA polyadenylation depends on the cytoplasmic polyadenylation binding protein 1 (CPEB1). Cpeb1 mRNA translation and protein levels are decreased in old oocytes. This decrease causes de-repression of Ccnb1 translation in quiescent oocytes, premature CDK1 activation, and accelerated reentry into meiosis. De-repression of Ccnb1 is corrected by Cpeb1 mRNA injection in old oocytes. Oocyte-specific Cpeb1 haploinsufficiency in young oocytes recapitulates all the translation phenotypes of old oocytes. These findings demonstrate that a dysfunction in the oocyte translation program is associated with the decline in oocyte quality during aging.
Collapse
Affiliation(s)
- Nozomi Takahashi
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Federica Franciosi
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,Reproductive and Developmental Biology Lab, Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 20133, Milan, Italy
| | - Enrico Maria Daldello
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Xuan G Luong
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Peter Althoff
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Xiaotian Wang
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Marco Conti
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA. .,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA. .,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
26
|
Jiang X, Xu X, Wang B, Song K, Zhang J, Chen Y, Tian Y, Weng J, Liang Y, Ma W. Adverse effects of 2-Methoxyestradiol on mouse oocytes during reproductive aging. Chem Biol Interact 2023; 369:110277. [PMID: 36414027 DOI: 10.1016/j.cbi.2022.110277] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/26/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
2-Methoxyestradiol (2-ME2) is a metabolite of 17β-estradiol and is currently in clinical trials as an antitumor agent. Here we found 2-ME2 level remains stable in the local environment of ovaries but declines in serum in aging mice, and exogenous 2-ME2 impacts the meiotic maturation of mouse oocytes in dose-dependent manner. In vitro 2-ME2 application arrested oocytes at metaphase I (MI), with abnormal spindle structure and chromosome alignment. 2-ME2 exposure induced excessive production of reactive oxygen species (ROS) and malondialdehyde, as well as accelerated apoptosis progression. 2-ME2 unbalanced mitochondrial dynamics by increasing DRP1 and MFN1 while decreasing Opa1. Similar phenotypes were also observed in oocytes from mice injected intraperitoneally with 2-ME2. Taken together, this study indicates 2-ME2 exposure impairs oocyte meiotic maturation through inducing mitochondrial imbalance, oxidative stress and apoptosis. The gradual decline in oocyte quality and quantity may be associated with the stable 2-ME2 in ovaries during female reproductive aging.
Collapse
Affiliation(s)
- Xiuying Jiang
- Devision of Sport Anatomy, School of Sport Science, Beijing Sport University, Beijing, 100084, China
| | - Xiangning Xu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Bicheng Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Ke Song
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jiaqi Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Ye Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Ying Tian
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jing Weng
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yuanjing Liang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wei Ma
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
27
|
Martin JH, Nixon B, Cafe SL, Aitken RJ, Bromfield EG, Lord T. OXIDATIVE STRESS AND REPRODUCTIVE FUNCTION: Oxidative stress and in vitro ageing of the post-ovulatory oocyte: an update on recent advances in the field. Reproduction 2022; 164:F109-F124. [PMID: 36190194 DOI: 10.1530/rep-22-0206] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/03/2022] [Indexed: 11/09/2022]
Abstract
In brief Post-ovulatory ageing of oocytes leads to poor oocyte and embryo quality as well as abnormalities in offspring. This review provides an update on the contributions of oxidative stress to this process and discusses the current literature surrounding the use of antioxidant media to delay post-ovulatory oocyte ageing. Abstract Following ovulation, the metaphase II stage oocyte has a limited functional lifespan before succumbing to a process known as post-ovulatory oocyte ageing. This progressive demise occurs both in vivo and in vitro and is accompanied by a deterioration in oocyte quality, leading to a well-defined sequelae of reduced fertilisation rates, poor embryo quality, post-implantation errors, and abnormalities in the offspring. Although the physiological consequences of post-ovulatory oocyte ageing have largely been characterised, less is known regarding the molecular mechanisms that drive this process. This review presents an update on the established relationships between the biochemical changes exhibited by the ageing oocyte and the myriad of symptoms associated with the ageing phenotype. In doing so, we consider the molecular events that are potentially involved in orchestrating post-ovulatory ageing with a particular focus on the role of oxidative stress. We highlight the mounting evidence that oxidative stress acts as an initiator for a cascade of events that create the aged oocyte phenotype. Specifically, oxidative stress has the capacity to disrupt mitochondrial function and directly damage multiple intracellular components of the oocyte such as lipids, proteins, and DNA. Finally, this review addresses emerging strategies for delaying post-ovulatory oocyte ageing with emphasis placed on the promise afforded by the use of selected antioxidants to guide the development of media tailored for the preservation of oocyte integrity during in vitro fertilisation procedures.
Collapse
Affiliation(s)
- Jacinta H Martin
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute Program in Infertility and Reproduction, New Lambton Heights, New South Wales, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute Program in Infertility and Reproduction, New Lambton Heights, New South Wales, Australia
| | - Shenae L Cafe
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute Program in Infertility and Reproduction, New Lambton Heights, New South Wales, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute Program in Infertility and Reproduction, New Lambton Heights, New South Wales, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute Program in Infertility and Reproduction, New Lambton Heights, New South Wales, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute Program in Infertility and Reproduction, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
28
|
Lodde V, Garcia Barros R, Terzaghi L, Franciosi F, Luciano AM. Insights on the Role of PGRMC1 in Mitotic and Meiotic Cell Division. Cancers (Basel) 2022; 14:cancers14235755. [PMID: 36497237 PMCID: PMC9736406 DOI: 10.3390/cancers14235755] [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: 09/11/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
During mitosis, chromosome missegregation and cytokinesis defects have been recognized as hallmarks of cancer cells. Cytoskeletal elements composing the spindle and the contractile ring and their associated proteins play crucial roles in the faithful progression of mitotic cell division. The hypothesis that PGRMC1, most likely as a part of a yet-to-be-defined complex, is involved in the regulation of spindle function and, more broadly, the cytoskeletal machinery driving cell division is particularly appealing. Nevertheless, more than ten years after the preliminary observation that PGRMC1 changes its localization dynamically during meiotic and mitotic cell division, this field of research has remained a niche and needs to be fully explored. To encourage research in this fascinating field, in this review, we will recap the current knowledge on PGRMC1 function during mitotic and meiotic cell division, critically highlighting the strengths and limitations of the experimental approaches used so far. We will focus on known interacting partners as well as new putative associated proteins that have recently arisen in the literature and that might support current as well as new hypotheses of a role for PGRMC1 in specific spindle subcompartments, such as the centrosome, kinetochores, and the midzone/midbody.
Collapse
|
29
|
Tetrabromobisphenol Exposure Impairs Bovine Oocyte Maturation by Inducing Mitochondrial Dysfunction. Molecules 2022; 27:molecules27228111. [PMID: 36432212 PMCID: PMC9696588 DOI: 10.3390/molecules27228111] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Tetrabromobisphenol (TBBPA) is the most widely used brominated flame retardant in the world and displays toxicity to humans and animals. However, few studies have focused on its impact on oocyte maturation. Here, TBBPA was added to the culture medium of bovine cumulus-oocyte complexes (COCs) to examine its effect on oocytes. We found that TBBPA exposure displayed an adverse influence on oocyte maturation and subsequent embryonic development. The results of this study showed that TBBPA exposure induced oocyte meiotic failure by disturbing the polar-body extrusion of oocytes and the expansion of cumulus cells. We further found that TBBPA exposure led to defective spindle assembly and chromosome alignment. Meanwhile, TBBPA induced oxidative stress and early apoptosis by mediating the expression of superoxide dismutase 2 (SOD2). TBBPA exposure also caused mitochondrial dysfunction, displaying a decrease in mitochondrial membrane potential, mitochondrial content, mtDNA copy number, and ATP levels, which are regulated by the expression of pyruvate dehydrogenase kinase 3 (PDK3). In addition, the developmental competence of oocytes and the quality of blastocysts were also reduced after TBBPA treatment. These results demonstrated that TBBPA exposure impaired oocyte maturation and developmental competence by disrupting both nuclear and cytoplasmic maturation of the oocyte, which might have been caused by oxidative stress induced by mitochondrial dysfunction.
Collapse
|
30
|
Wu T, Gu H, Luo Y, Wang L, Sang Q. Meiotic defects in human oocytes: Potential causes and clinical implications. Bioessays 2022; 44:e2200135. [PMID: 36207289 DOI: 10.1002/bies.202200135] [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: 07/07/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/09/2022]
Abstract
Meiotic defects cause abnormal chromosome segregation leading to aneuploidy in mammalian oocytes. Chromosome segregation is particularly error-prone in human oocytes, but the mechanisms behind such errors remain unclear. To explain the frequent chromosome segregation errors, recent investigations have identified multiple meiotic defects and explained how these defects occur in female meiosis. In particular, we review the causes of cohesin exhaustion, leaky spindle assembly checkpoint (SAC), inherently unstable meiotic spindle, fragmented kinetochores or centromeres, abnormal aurora kinases (AURK), and clinical genetic variants in human oocytes. We mainly focus on meiotic defects in human oocytes, but also refer to the potential defects of female meiosis in mouse models.
Collapse
Affiliation(s)
- Tianyu Wu
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Hao Gu
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Yuxi Luo
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Abstract
A hormone enhances cognition in mouse models of Alzheimer's disease and Down syndrome.
Collapse
Affiliation(s)
- Hanne M Hoffmann
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
32
|
Leem J, Kim S, Kim JS, Oh JS. ROS-independent cytotoxicity of 9,10-phenanthrenequinone inhibits cell cycle progression and spindle assembly during meiotic maturation in mouse oocytes. JOURNAL OF HAZARDOUS MATERIALS 2022; 436:129248. [PMID: 35739767 DOI: 10.1016/j.jhazmat.2022.129248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/10/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Diesel exhaust particles (DEPs) are major components of ambient particulate matter and are associated with various adverse health effects. Typically, DEPs contain a vast number of organic compounds, among which 9,10-phenanthrenequinone (9,10-PQ), the quinone derivative of the polycyclic aromatic hydrocarbon phenanthrene, is one of the most abundant and toxic. 9,10-PQ can produce excessive reactive oxygen species (ROS) via redox cycling and exhibit cytotoxicity in various cells. However, the underlying mechanisms involved in cytotoxicity of 9,10-PQ remain elusive. In this study, we investigated the effects of exposure to 9,10-PQ using mouse oocytes as a model system. We found that 9,10-PQ compromised meiotic maturation by impairing acentriolar microtubule organizing center (MTOC) assembly and subsequent spindle formation during meiotic maturation. Moreover, 9,10-PQ exposure prevented cell cycle progression by inhibiting Cdk1 activation via disturbance of cyclin B1 accumulation. Importantly, meiotic defects induced by 9,10-PQ exposure were not rescued by decreasing ROS levels, revealing that 9,10-PQ has ROS-independent activity that regulates cell cycle progression and spindle assembly. Therefore, our findings reveal that 9,10-PQ has novel activity that regulates cell-cycle progression and spindle formation in an ROS-independent manner during meiotic maturation in mouse oocytes.
Collapse
Affiliation(s)
- Jiyeon Leem
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Seul Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jae-Sung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jeong Su Oh
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
33
|
Meng TG, Lei WL, Lu X, Liu XY, Ma XS, Nie XQ, Zhao ZH, Li QN, Huang L, Hou Y, Ouyang YC, Li L, Tang TS, Schatten H, Xie W, Gao SR, Ou XH, Wang ZB, Sun QY. Maternal EHMT2 is essential for homologous chromosome segregation by regulating Cyclin B3 transcription in oocyte meiosis. Int J Biol Sci 2022; 18:4513-4531. [PMID: 35864958 PMCID: PMC9295060 DOI: 10.7150/ijbs.75298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/29/2022] [Indexed: 11/05/2022] Open
Abstract
During oocyte growth, various epigenetic modifications are gradually established, accompanied by accumulation of large amounts of mRNAs and proteins. However, little is known about the relationship between epigenetic modifications and meiotic progression. Here, by using Gdf9-Cre to achieve oocyte-specific ablation of Ehmt2 (Euchromatic-Histone-Lysine-Methyltransferase 2) from the primordial follicle stage, we found that female mutant mice were infertile. Oocyte-specific knockout of Ehmt2 caused failure of homologous chromosome separation independent of persistently activated SAC during the first meiosis. Further studies revealed that lacking maternal Ehmt2 affected the transcriptional level of Ccnb3, while microinjection of exogenous Ccnb3 mRNA could partly rescue the failure of homologous chromosome segregation. Of particular importance was that EHMT2 regulated ccnb3 transcriptions by regulating CTCF binding near ccnb3 gene body in genome in oocytes. In addition, the mRNA level of Ccnb3 significantly decreased in the follicles microinjected with Ctcf siRNA. Therefore, our findings highlight the novel function of maternal EHMT2 on the metaphase I-to-anaphase I transition in mouse oocytes: regulating the transcription of Ccnb3.
Collapse
Affiliation(s)
- Tie-Gang Meng
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wen-Long Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xukun Lu
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.,Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiao-Yu Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
| | - Xue-Shan Ma
- The Affiliated Tai'an City Central Hospital of Qingdao University, Taian, Shandong, 271000, China
| | - Xiao-Qing Nie
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zheng-Hui Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100101, China
| | - Qian-Nan Li
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lin Huang
- Center for Clinical Medicine Research, The Affiliated Hospital of Southwest Medical University, Luzhou 6460000, China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Tie-Shan Tang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Wei Xie
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.,Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shao-Rong Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
| | - Xiang-Hong Ou
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100101, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
34
|
Abstract
Many human embryos die in utero owing to an excess or deficit of chromosomes, a phenomenon known as aneuploidy; this is largely a consequence of nondisjunction during maternal meiosis I. Asymmetries of this division render it vulnerable to selfish centromeres that promote their own transmission, these being thought to somehow underpin aneuploidy. In this essay, I suggest that these vulnerabilities provide only half the solution to the enigma. In mammals, as in utero and postnatal provisioning is continuous, the costs of early death are mitigated. With such reproductive compensation, selection can favour a centromere because it induces lethal aneuploidy: if, when taken towards the polar body, it instead kills the embryo via aneuploidy, it gains. The model is consistent with the observation that reduced dosage of a murine drive suppressor induces aneuploidy and with the fact that high aneuploidy rates in vertebrates are seen exclusively in mammals. I propose further tests of this idea. The wastefulness of human reproduction may be a price we pay for nurturing our offspring.
Collapse
Affiliation(s)
- Laurence D. Hurst
- Wissenshaftskolleg zu Berlin, Berlin, Germany
- The Milner Centre for Evolution, University of Bath, Bath, Somerset, United Kingdom
| |
Collapse
|
35
|
Park S, Jeon HJ, Choi DY, Oh JS. Polystyrene nanoparticles incorporate into the endoplasmic reticulum and disturb translation during meiotic maturation in mouse oocytes. Toxicol In Vitro 2022; 82:105380. [PMID: 35561955 DOI: 10.1016/j.tiv.2022.105380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/20/2022] [Accepted: 05/06/2022] [Indexed: 12/06/2022]
Abstract
Polystyrene (PS) is one of the most common polymers that cause plastic pollution after release into the environment. Although a growing body of evidence has shown the adverse effects of PS on living organisms including humans, their effects on mammalian oocytes have not been extensively studied. In this study, we investigated the effect of exposure to PS-nanoparticle (PS-NPs) on meiotic maturation in mouse oocytes. We found that exogenous PS-NPs internalized into oocytes after penetrating the zona pellucida and accumulated in the cytoplasm of oocytes during meiotic maturation. Exposure to PS-NPs did not affect meiotic resumption but inhibited meiotic maturation by impairing spindle assembly and chromosome alignment. Moreover, exposure to PS-NPs increased oxidative stress and mitochondrial aggregation during meiotic maturation. Notably, internalized PS-NPs localized around the endoplasmic reticulum (ER) and disturbed translation in oocytes. Therefore, it is suggested that PS-NPs impair meiotic maturation not only by increasing oxidative stress and mitochondrial dysfunction, but also by decreasing translation efficiency after incorporating into the ER during meiotic maturation in mouse oocytes.
Collapse
Affiliation(s)
- Seongwoo Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hyuk-Joon Jeon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Da Yi Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jeong Su Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
36
|
Chromosome Segregation in the Oocyte: What Goes Wrong during Aging. Int J Mol Sci 2022; 23:ijms23052880. [PMID: 35270022 PMCID: PMC8911062 DOI: 10.3390/ijms23052880] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/22/2022] [Accepted: 03/05/2022] [Indexed: 12/13/2022] Open
Abstract
Human female fertility and reproductive lifespan decrease significantly with age, resulting in an extended post-reproductive period. The central dogma in human female reproduction contains two important aspects. One is the pool of oocytes in the human ovary (the ovarian reserve; approximately 106 at birth), which diminishes throughout life until menopause around the age of 50 (approximately 103 oocytes) in women. The second is the quality of oocytes, including the correctness of meiotic divisions, among other factors. Notably, the increased rate of sub- and infertility, aneuploidy, miscarriages, and birth defects are associated with advanced maternal age, especially in women above 35 years of age. This postponement is also relevant for human evolution; decades ago, the female aging-related fertility drop was not as important as it is today because women were having their children at a younger age. Spindle assembly is crucial for chromosome segregation during each cell division and oocyte maturation, making it an important event for euploidy. Consequently, aberrations in this segregation process, especially during the first meiotic division in human eggs, can lead to implantation failure or spontaneous abortion. Today, human reproductive medicine is also facing a high prevalence of aneuploidy, even in young females. However, the shift in the reproductive phase of humans and the strong increase in errors make the problem much more dramatic at later stages of the female reproductive phase. Aneuploidy in human eggs could be the result of the non-disjunction of entire chromosomes or sister chromatids during oocyte meiosis, but partial or segmental aneuploidies are also relevant. In this review, we intend to describe the relevance of the spindle apparatus during oocyte maturation for proper chromosome segregation in the context of maternal aging and the female reproductive lifespan.
Collapse
|
37
|
Ozturk S. Molecular determinants of the meiotic arrests in mammalian oocytes at different stages of maturation. Cell Cycle 2022; 21:547-571. [PMID: 35072590 PMCID: PMC8942507 DOI: 10.1080/15384101.2022.2026704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/21/2021] [Accepted: 01/03/2022] [Indexed: 01/26/2023] Open
Abstract
Mammalian oocytes undergo two rounds of developmental arrest during maturation: at the diplotene of the first meiotic prophase and metaphase of the second meiosis. These arrests are strictly regulated by follicular cells temporally producing the secondary messengers, cAMP and cGMP, and other factors to regulate maturation promoting factor (composed of cyclin B1 and cyclin-dependent kinase 1) levels in the oocytes. Out of these normally appearing developmental arrests, permanent arrests may occur in the oocytes at germinal vesicle (GV), metaphase I (MI), or metaphase II (MII) stage. This issue may arise from absence or altered expression of the oocyte-related genes playing key roles in nuclear and cytoplasmic maturation. Additionally, the assisted reproductive technology (ART) applications such as ovarian stimulation and in vitro culture conditions both of which harbor various types of chemical agents may contribute to forming the permanent arrests. In this review, the molecular determinants of developmental and permanent arrests occurring in the mammalian oocytes are comprehensively evaluated in the light of current knowledge. As number of permanently arrested oocytes at different stages is increasing in ART centers, potential approaches for inducing permanent arrests to obtain competent oocytes are discussed.
Collapse
Affiliation(s)
- Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
38
|
Yue W, Wang Y, Meng T, Zhang H, Zhang X, Ouyang Y, Hou Y, Schatten H, Wang Z, Sun Q. Kinetochore scaffold 1 regulates SAC function during mouse oocyte meiotic maturation. FASEB J 2022; 36:e22210. [DOI: 10.1096/fj.202101586rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Wei Yue
- 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
| | - Yue Wang
- College of Animal Science and Technology Nanjing Agricultural University Nanjing China
| | - Tie‐Gang Meng
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
| | - Hong‐Yong Zhang
- Department of Reproductive Medicine Peking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical Center Shenzhen China
| | - Xin‐Ran Zhang
- 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
| | - Ying‐Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
| | - Heide Schatten
- Department of Veterinary Pathobiology University of Missouri Columbia Missouri USA
| | - Zhen‐Bo Wang
- 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
| | - Qing‐Yuan Sun
- Fertility Preservation Lab, Guangdong‐Hong Kong Metabolism & Reproduction Joint Laboratory Reproductive Medicine Center Guangdong Second Provincial General Hospital Guangzhou China
| |
Collapse
|
39
|
Wang YS, Chen C, Ahmad MJ, Chen F, Ding ZM, Yang SJ, Chen YW, Duan ZQ, Liu M, Liang AX, He CJ, Hua GH, Huo LJ. WDR62 regulates mouse oocyte meiotic maturation related to p-JNK and H3K9 trimethylation. Int J Biochem Cell Biol 2022; 144:106169. [PMID: 35093571 DOI: 10.1016/j.biocel.2022.106169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 01/01/2023]
Abstract
WDR62 (WD40-repeat protein 62) participates in diverse biological process, especially mitotic spindle organization via regulating centriole biogenesis and the function of centriole-associated protein. However, the role of WDR62 exerts in spindle assembly and meiotic progression control in oocytes lacking typical centrosomes remains obscure. In a previous study, we reported that WDR62 is involved in spindle migration and asymmetric cytokinesis in mouse oocyte meiosis. In the current study, another novel function of WDR62 regulating cell cycle progression through meiotic spindle formation during oocyte meiotic maturation was found. Knockdown of WDR62 through siRNA microinjection disrupted the meiotic cell cycle and induced metaphase-I (MI) arrest coupled with severe spindle abnormality, chromosome misalignment, and aneuploid generation. Moreover, WDR62 depletion induced defective kinetochore-microtubule attachments (K-MT) and activated spindle assembly checkpoint (SAC), which could trigger the arrest of meiotic progression. Further study demonstrated that depletion of WDR62 was associated with an aberrant location of p-JNK and reduced its expression level; concomitantly, status of H3K9 trimethylation was also altered. In addition, phenotypes similar to WDR62 depletion were observed during the function-loss analysis of p-JNK using a specific inhibitor (SP600125), which signifies that WDR62 is important for spindle organization and meiotic progression, and this function might be via its regulation of p-JNK. In conclusion, this study revealed that WDR62 functions in multiple ways during oocyte meiotic maturation, which could be related to p-JNK and H3K9 trimethylation.
Collapse
Affiliation(s)
- Yong-Sheng Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Chao Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Muhammad Jamil Ahmad
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Fan Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Zhi-Ming Ding
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Sheng-Ji Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Yang-Wu Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Ze-Qun Duan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Ming Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Ai-Xin Liang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Chang-Jiu He
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Guo-Hua Hua
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Li-Jun Huo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| |
Collapse
|
40
|
Shimoi G, Wakabayashi R, Ishikawa R, Kameyama Y. Effects of post-ovulatory aging on centromeric cohesin protection in murine MII oocytes. Reprod Med Biol 2022; 21:RMB212433. [PMID: 35386382 PMCID: PMC8967304 DOI: 10.1002/rmb2.12433] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 10/30/2021] [Accepted: 11/23/2021] [Indexed: 11/09/2022] Open
Abstract
Purpose Post-ovulatory aging causes a high frequency of aneuploidy during meiosis II in mouse oocytes, irrespective of maternal age. In this study, we evaluated the effects of post-ovulatory oocyte aging on the protection of chromosomal cohesion involved in aneuploidy and verified the relationship between PP2A or SGO2 expression and the phosphorylation level of REC8 in oocytes. Methods Murine ovulated oocytes were incubated for 6 or 12 h in vitro after collection and denoted as the aged group. The oocytes examined immediately after collection were used as the control group. Immunofluorescent staining was used to detect the localization of PP2A, SGO2, BUB1, AURORA B, and MAD2 in the chromosomal centromere. Immunoblotting was used to quantify the expression of proteins describe above and REC8 in the oocytes. Results PP2A expression involved in the de-phosphorylation of REC8 decreased over time in oocytes, suggesting a deficiency in PP2A in centromeres. This indicated an increase in the level of phosphorylated REC8, which destabilizes centromeric cohesion in oocytes. In contrast, SGO2 expression was significantly high in aged oocytes. Conclusions The findings show that post-ovulatory aging destabilizes the centromeric cohesin protection in oocytes and can cause aneuploidy, which is often observed in aged oocytes during meiosis II.
Collapse
Affiliation(s)
- Gaku Shimoi
- Faculty of BioindustryTokyo University of AgricultureAbashiriJapan
- Graduate School of BioindustryTokyo University of AgricultureAbashiriJapan
| | - Rico Wakabayashi
- Faculty of BioindustryTokyo University of AgricultureAbashiriJapan
| | - Ryu Ishikawa
- Graduate School of BioindustryTokyo University of AgricultureAbashiriJapan
| | - Yuichi Kameyama
- Faculty of BioindustryTokyo University of AgricultureAbashiriJapan
- Graduate School of BioindustryTokyo University of AgricultureAbashiriJapan
| |
Collapse
|
41
|
Buratini J, Dellaqua TT, Dal Canto M, La Marca A, Carone D, Mignini Renzini M, Webb R. The putative roles of FSH and AMH in the regulation of oocyte developmental competence: from fertility prognosis to mechanisms underlying age-related subfertility. Hum Reprod Update 2021; 28:232-254. [PMID: 34969065 DOI: 10.1093/humupd/dmab044] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 11/18/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Fertility loss during female ageing is associated with increasing basal FSH and decreasing anti-Müllerian hormone (AMH) concentrations, together with compromised oocyte quality, presumably due to increased oxidative stress (OS) and DNA damage, as well as reduced metabolic and meiotic competences. Basal FSH and AMH circulatory concentrations have been broadly utilized as IVF success predictors, regardless of fluctuations in prognostic accuracy; basal FSH and AMH perform better in pre-advanced maternal age (AMA: >35 years) and AMA patients, respectively. The relationships between FSH and AMH intrafollicular levels and IVF outcomes suggest, nevertheless, that both hormones regulate oocyte competence, supporting the hypothesis that changes in FSH/AMH levels cause, at least in part, oocyte quality degradation during ageing. To understand the reasons behind the fluctuations in FSH and AMH prognostic accuracies and to clarify their participation in mechanisms determining oocyte competence and age-related subfertility, a deeper knowledge of the regulation of FSH and AMH intrafollicular signalling during the female reproductive lifespan, and of their effects on the cumulus-oocyte complex, is required. OBJECTIVE AND RATIONALE An extensive body of information on the regulation of FSH and AMH intrafollicular availability and signalling, as well as on the control of folliculogenesis and oocyte metabolism, has been accumulated. However, these datasets have been explored within the relatively narrow boundaries of their specific subjects. Given the aforementioned gaps in knowledge and their clinical relevance, herein we integrate clinical and basic data, within a wide biological perspective, aiming to shed light on (i) the reasons for the variability in the accuracy of serum FSH and AMH as fertility markers, and on (ii) the potential roles of these hormones in mechanisms regulating oocyte quality, particularly those associated with ageing. SEARCH METHODS The PubMed database encompassing the period between 1960 and 2021 was searched. Principal search terms were FSH, FSH receptor, AMH, oocyte, maternal age, cumulus, transzonal projections (TZPs), actin, OS, redox, reactive oxygen species, mitochondria, DNA damage, DNA repair, aneuploidy, spindle, meiosis, gene expression, transcription, translation, oocyte secreted factors (OSFs), cAMP, cyclic guanosine monophosphate, natriuretic peptide C, growth differentiation factor 9, bone morphogenetic protein 15 and fibroblast growth factor. OUTCOMES Our analysis suggests that variations in the accuracy of fertility prognosis reflect a modest association between circulatory AMH levels and oocyte quality as well as increasing basal FSH inter-cycle variability with age. In addition, the basic and clinical data articulated herein support the hypothesis that increased intrafollicular FSH levels, as maternal age advances, may override the physiological protective influences of AMH and OSFs against excessive FSH signalling in cumulus cells. This would result in the disruption of oocyte homeostasis via reduced TZP-mediated transfer of cumulus-derived molecules essential for meiotic competence, gene expression, redox activity and DNA repair. WIDER IMPLICATIONS In-depth data analysis, encompassing a wide biological perspective has revealed potential causative mechanisms of age-related subfertility triggered by alterations in FSH/AMH signalling during the female reproductive life. Insights from new mechanistic models arising from this analysis should contribute to advancing our comprehension of oocyte biology in humans and serve as a valuable reference for novel AMA subfertility treatments aimed at improving oocyte quality through the modulation of AMH/FSH action.
Collapse
Affiliation(s)
- Jose Buratini
- Biogenesi Reproductive Medicine Centre-Eugin Group, Istituti Clinici Zucchi, Monza, Italy.,Clinica Eugin Modena, Modena, Italy.,Department of Structural and Functional Biology, Sao Paulo State University, Botucatu, Brazil
| | - Thaisy Tino Dellaqua
- Department of Structural and Functional Biology, Sao Paulo State University, Botucatu, Brazil
| | - Mariabeatrice Dal Canto
- Biogenesi Reproductive Medicine Centre-Eugin Group, Istituti Clinici Zucchi, Monza, Italy.,Clinica Eugin Modena, Modena, Italy
| | - Antonio La Marca
- Clinica Eugin Modena, Modena, Italy.,Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Mario Mignini Renzini
- Biogenesi Reproductive Medicine Centre-Eugin Group, Istituti Clinici Zucchi, Monza, Italy.,Clinica Eugin Modena, Modena, Italy
| | - Robert Webb
- Division of Animal Sciences, School of Biosciences, University of Nottingham, Nottinghamshire, UK
| |
Collapse
|
42
|
Mercer M, Jang S, Ni C, Buszczak M. The Dynamic Regulation of mRNA Translation and Ribosome Biogenesis During Germ Cell Development and Reproductive Aging. Front Cell Dev Biol 2021; 9:710186. [PMID: 34805139 PMCID: PMC8595405 DOI: 10.3389/fcell.2021.710186] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/07/2021] [Indexed: 01/21/2023] Open
Abstract
The regulation of mRNA translation, both globally and at the level of individual transcripts, plays a central role in the development and function of germ cells across species. Genetic studies using flies, worms, zebrafish and mice have highlighted the importance of specific RNA binding proteins in driving various aspects of germ cell formation and function. Many of these mRNA binding proteins, including Pumilio, Nanos, Vasa and Dazl have been conserved through evolution, specifically mark germ cells, and carry out similar functions across species. These proteins typically influence mRNA translation by binding to specific elements within the 3′ untranslated region (UTR) of target messages. Emerging evidence indicates that the global regulation of mRNA translation also plays an important role in germ cell development. For example, ribosome biogenesis is often regulated in a stage specific manner during gametogenesis. Moreover, oocytes need to produce and store a sufficient number of ribosomes to support the development of the early embryo until the initiation of zygotic transcription. Accumulating evidence indicates that disruption of mRNA translation regulatory mechanisms likely contributes to infertility and reproductive aging in humans. These findings highlight the importance of gaining further insights into the mechanisms that control mRNA translation within germ cells. Future work in this area will likely have important impacts beyond germ cell biology.
Collapse
Affiliation(s)
- Marianne Mercer
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Seoyeon Jang
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chunyang Ni
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Michael Buszczak
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
43
|
Li L, Xia Y, Yang Y, Zhang W, Yan H, Yin P, Li K, Chen Y, Lu L, Tong G. CDC26 is a key factor in human oocyte aging. Hum Reprod 2021; 36:3095-3107. [PMID: 34590680 DOI: 10.1093/humrep/deab217] [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: 01/13/2021] [Revised: 08/01/2021] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Is CDC26 a key factor in human oocyte aging? SUMMARY ANSWER The lack of CDC26 disrupts the oocytes maturation process, leading to oocyte aging, but these defects could be partially rescued by overexpression of the CDC26 protein. WHAT IS KNOWN ALREADY Age-related oocyte aging is the main cause of female fertility decline. In mammalian oocytes, aberrant meiosis can cause chromosomal abnormalities that might lead to infertility and developmental disorders. CDC26 participates in the meiosis process. STUDY DESIGN, SIZE, DURATION Differential gene expression in young and old women oocytes were screened by single-cell RNA-seq technology, and the functions of differentially genes were verified on mouse oocytes. Finally, transfection technology was used to evaluate the effect of a differentially expressed gene in rescuing human oocyte from aging. PARTICIPANTS/MATERIALS, SETTING, METHODS Discarded human oocytes were collected for single-cell RNA-seq, q-PCR and immunocytochemical analyses to screen for and identify differential gene expression. Female KM mice oocytes were collected for IVM of oocytes, q-PCR and immunocytochemical analyses to delineate the relationships between oocyte aging and differential gene expression. Additionally, recombinant lentiviral vectors encoding CDC26 were transfected into the germinal vesicle oocytes of older women, to investigate the effects of the CDC26 gene expression on oocyte development. MAIN RESULTS AND THE ROLE OF CHANCE Many genes were found to be differentially expressed in the oocytes of young versus old patients via RNA-seq technology. CDC26 mRNA and protein levels in aged oocytes were severely decreased, when compared with the levels observed in young oocytes. Moreover, aged oocytes lacking CDC26 were more prone to aneuploidy. These defects in aged oocytes could be partially rescued by overexpression of the CDC26 protein. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Our study delineated key steps in the oocyte aging process by identifying the key role of CDC26 in the progression of oocyte maturation. Future studies are required to address whether other signaling pathways play a role in regulating oocyte maturation via CDC26 and which genes are the direct molecular targets of CDC26. WIDER IMPLICATIONS OF THE FINDINGS Our results using in vitro systems for both mouse and human oocyte maturation provide a proof of principle that CDC26 may represent a novel therapeutic approach against maternal aging-related spindle and chromosomal abnormalities. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from the National Natural Science Foundation of China (81571442 and 81170571), the outstanding Talent Project of Shanghai Municipal Commission of Health (XBR2011067) and Clinical Research and Cultivation Project in Shanghai Municipal Hospitals (SHDC12019X32). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Li Li
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ye Xia
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Yang
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wuwen Zhang
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Yan
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Yin
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kai Li
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Chen
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Lu
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guoqing Tong
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
44
|
Nabi D, Drechsler H, Pschirer J, Korn F, Schuler N, Diez S, Jessberger R, Chacón M. CENP-V is required for proper chromosome segregation through interaction with spindle microtubules in mouse oocytes. Nat Commun 2021; 12:6547. [PMID: 34764261 PMCID: PMC8586017 DOI: 10.1038/s41467-021-26826-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 10/21/2021] [Indexed: 11/08/2022] Open
Abstract
Proper chromosome segregation is essential to avoid aneuploidy, yet this process fails with increasing age in mammalian oocytes. Here we report a role for the scarcely described protein CENP-V in oocyte spindle formation and chromosome segregation. We show that depending on the oocyte maturation state, CENP-V localizes to centromeres, to microtubule organizing centers, and to spindle microtubules. We find that Cenp-V-/- oocytes feature severe deficiencies, including metaphase I arrest, strongly reduced polar body extrusion, increased numbers of mis-aligned chromosomes and aneuploidy, multipolar spindles, unfocused spindle poles and loss of kinetochore spindle fibres. We also show that CENP-V protein binds, diffuses along, and bundles microtubules in vitro. The spindle assembly checkpoint arrests about half of metaphase I Cenp-V-/- oocytes from young adults only. This finding suggests checkpoint weakening in ageing oocytes, which mature despite carrying mis-aligned chromosomes. Thus, CENP-V is a microtubule bundling protein crucial to faithful oocyte meiosis, and Cenp-V-/- oocytes reveal age-dependent weakening of the spindle assembly checkpoint.
Collapse
Affiliation(s)
- Dalileh Nabi
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Neuropediatrics Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Hauke Drechsler
- B CUBE-Center for Molecular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Johannes Pschirer
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Franz Korn
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nadine Schuler
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan Diez
- B CUBE-Center for Molecular Bioengineering, Technische Universität Dresden, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Rolf Jessberger
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | - Mariola Chacón
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- CABIMER, Centro Andaluz de Biología Molecular & Medicina Regenerativa, Sevilla, Spain.
| |
Collapse
|
45
|
Zhou C, Zhang X, Miao Y, Zhang Y, Li Y, Xiong B. The cohesin stabilizer Sororin drives G 2-M transition and spindle assembly in mammalian oocytes. SCIENCE ADVANCES 2021; 7:eabg9335. [PMID: 34559563 PMCID: PMC8462903 DOI: 10.1126/sciadv.abg9335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
During the S phase of mitosis, Sororin is recruited by acetylated Smc3 and stabilizes sister chromatid cohesion by counteracting the Wapl-Pds5 interaction. Thereafter, Sororin is phosphorylated during prophase and translocated to the cytoplasm, where its function remains poorly understood. Here, we report that Sororin acts as a regulator of meiotic G2-M transition and spindle assembly in mammalian oocytes. Sororin is present in the nucleus of GV oocytes and becomes associated with the spindle apparatus during meiosis I in mice. Depletion of Sororin causes failure of GVBD due to inactivation of Cdk1 and defective spindle assembly because of reduced levels of Cyclin B2. We validate Sororin interactions with Cyclin B2 that protects it from destruction by APCCdh1, which drives M phase entry and bipolar spindle formation. Notably, the meiotic functions of Sororin are conserved among mammals. Together, our findings provide novel insights into the noncanonical role of Sororin in the resumption of meiosis and progression through meiosis I in mammalian oocytes.
Collapse
|
46
|
Wu T, Lane SIR, Morgan SL, Tang F, Jones KT. Loss of centromeric RNA activates the spindle assembly checkpoint in mammalian female meiosis I. J Cell Biol 2021; 220:212548. [PMID: 34379093 PMCID: PMC8360762 DOI: 10.1083/jcb.202011153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 05/01/2021] [Accepted: 07/27/2021] [Indexed: 12/22/2022] Open
Abstract
The repetitive sequences of DNA centromeric regions form the structural basis for kinetochore assembly. Recently they were found to be transcriptionally active in mitosis, with their RNAs providing noncoding functions. Here we explore the role, in mouse oocytes, of transcripts generated from within the minor satellite repeats. Depletion of minor satellite transcripts delayed progression through meiosis I by activation of the spindle assembly checkpoint. Arrested oocytes had poorly congressed chromosomes, and centromeres were frequently split by microtubules. Thus, we have demonstrated that the centromeric RNA plays a specific role in female meiosis I compared with mitosis and is required for maintaining the structural integrity of centromeres. This may contribute to the high aneuploidy rates observed in female meiosis.
Collapse
Affiliation(s)
- Tianyu Wu
- Department of Central Laboratory, Clinical Laboratory, Jing'an District Centre Hospital of Shanghai, Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism and Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Simon I R Lane
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - Stephanie L Morgan
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Feng Tang
- Discipline of Obstetrics and Gynecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Keith T Jones
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| |
Collapse
|
47
|
Mishina T, Tabata N, Hayashi T, Yoshimura M, Umeda M, Mori M, Ikawa Y, Hamada H, Nikaido I, Kitajima TS. Single-oocyte transcriptome analysis reveals aging-associated effects influenced by life stage and calorie restriction. Aging Cell 2021; 20:e13428. [PMID: 34245092 PMCID: PMC8373347 DOI: 10.1111/acel.13428] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
Chromosome segregation errors in oocytes lead to the production of aneuploid eggs, which are the leading cause of pregnancy loss and of several congenital diseases such as Down syndrome. The frequency of chromosome segregation errors in oocytes increases with maternal age, especially at a late stage of reproductive life. How aging at various life stages affects oocytes differently remains poorly understood. In this study, we describe aging‐associated changes in the transcriptome profile of mouse oocytes throughout reproductive life. Our single‐oocyte comprehensive RNA sequencing using RamDA‐seq revealed that oocytes undergo transcriptome changes at a late reproductive stage, whereas their surrounding cumulus cells exhibit transcriptome changes at an earlier stage. Calorie restriction, a paradigm that reportedly prevents aging‐associated egg aneuploidy, promotes a transcriptome shift in oocytes with the up‐regulation of genes involved in chromosome segregation. This shift is accompanied by the improved maintenance of chromosomal cohesin, the loss of which is a hallmark of oocyte aging and causes chromosome segregation errors. These findings have implications for understanding how oocytes undergo aging‐associated functional decline throughout their reproductive life in a context‐dependent manner.
Collapse
Affiliation(s)
- Tappei Mishina
- Laboratory for Chromosome Segregation RIKEN Center for Biosystems Dynamics Research (BDR) Kobe Japan
| | - Namine Tabata
- Laboratory for Chromosome Segregation RIKEN Center for Biosystems Dynamics Research (BDR) Kobe Japan
- Graduate School of Biostudies Kyoto University Kyoto Japan
| | - Tetsutaro Hayashi
- Laboratory for Bioinformatics Research RIKEN Center for Biosystems Dynamics Research (BDR) Kobe Japan
| | - Mika Yoshimura
- Laboratory for Bioinformatics Research RIKEN Center for Biosystems Dynamics Research (BDR) Kobe Japan
| | - Mana Umeda
- Laboratory for Bioinformatics Research RIKEN Center for Biosystems Dynamics Research (BDR) Kobe Japan
| | - Masashi Mori
- Laboratory for Chromosome Segregation RIKEN Center for Biosystems Dynamics Research (BDR) Kobe Japan
| | - Yayoi Ikawa
- Laboratory for Organismal Patterning RIKEN Center for Biosystems Dynamics Research (BDR) Kobe Japan
| | - Hiroshi Hamada
- Laboratory for Organismal Patterning RIKEN Center for Biosystems Dynamics Research (BDR) Kobe Japan
| | - Itoshi Nikaido
- Laboratory for Bioinformatics Research RIKEN Center for Biosystems Dynamics Research (BDR) Kobe Japan
- Department of Functional Genome Informatics, Division of Medical Genomics, Medical Research Institute Tokyo Medical and Dental University (TMDU) Bunkyo Japan
- Master's/Doctoral Program in Life Science Innovation (Bioinformatics), Degree Programs in Systems and Information Engineering, Graduate School of Science and Technology University of Tsukuba Tsukuba Japan
| | - Tomoya S. Kitajima
- Laboratory for Chromosome Segregation RIKEN Center for Biosystems Dynamics Research (BDR) Kobe Japan
- Graduate School of Biostudies Kyoto University Kyoto Japan
| |
Collapse
|
48
|
Wang X, Baumann C, De La Fuente R, Viveiros MM. Loss of acentriolar MTOCs disrupts spindle pole Aurora A and assembly of the liquid-like meiotic spindle domain in oocytes. J Cell Sci 2021; 134:jcs256297. [PMID: 34152366 PMCID: PMC8325960 DOI: 10.1242/jcs.256297] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 06/14/2021] [Indexed: 11/20/2022] Open
Abstract
Oocyte-specific knockdown of pericentrin (PCNT) in transgenic (Tg) mice disrupts acentriolar microtubule-organizing center (aMTOC) formation, leading to spindle instability and error-prone meiotic division. Here, we show that PCNT-depleted oocytes lack phosphorylated Aurora A (pAURKA) at spindle poles, while overall levels are unaltered. To test aMTOC-associated AURKA function, metaphase II (MII) control (WT) and Tg oocytes were briefly exposed to a specific AURKA inhibitor (MLN8237). Similar defects were observed in Tg and MLN8237-treated WT oocytes, including altered spindle structure, increased chromosome misalignment and impaired microtubule regrowth. Yet, AURKA inhibition had a limited effect on Tg oocytes, revealing a critical role for aMTOC-associated AURKA in regulating spindle stability. Notably, spindle instability was associated with disrupted γ-tubulin and lack of the liquid-like meiotic spindle domain (LISD) in Tg oocytes. Analysis of this Tg model provides the first evidence that LISD assembly depends expressly on aMTOC-associated AURKA, and that Ran-mediated spindle formation ensues without the LISD. These data support that loss of aMTOC-associated AURKA and failure of LISD assembly contribute to error-prone meiotic division in PCNT-depleted oocytes, underscoring the essential role of aMTOCs for spindle stability.
Collapse
Affiliation(s)
- Xiaotian Wang
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia,Athens, GA 30602, USA
| | - Claudia Baumann
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia,Athens, GA 30602, USA
| | - Rabindranath De La Fuente
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia,Athens, GA 30602, USA
- Regenerative Biosciences Center (RBC), University of Georgia,Athens, GA 30602, USA
| | - Maria M. Viveiros
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia,Athens, GA 30602, USA
- Regenerative Biosciences Center (RBC), University of Georgia,Athens, GA 30602, USA
| |
Collapse
|
49
|
Advanced maternal age perturbs mouse embryo development and alters the phenotype of derived embryonic stem cells. J Dev Orig Health Dis 2021; 13:395-405. [PMID: 34193331 DOI: 10.1017/s2040174421000325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Advanced maternal age (AMA) is known to reduce fertility, increases aneuploidy in oocytes and early embryos and leads to adverse developmental consequences which may associate with offspring lifetime health risks. However, investigating underlying effects of AMA on embryo developmental potential is confounded by the inherent senescence present in maternal body systems further affecting reproductive success. Here, we describe a new model for the analysis of early developmental mechanisms underlying AMA by the derivation and characterisation of mouse embryonic stem cell (mESC-like) lines from naturally conceived embryos. Young (7-8 weeks) and Old (7-8 months) C57BL/6 female mice were mated with young males. Preimplantation embryos from Old dams displayed developmental retardation in blastocyst morphogenesis. mESC lines established from these blastocysts using conventional techniques revealed differences in genetic, cellular and molecular criteria conserved over several passages in the standardised medium. mESCs from embryos from AMA dams displayed increased incidence of aneuploidy following Giemsa karyotyping compared with those from Young dams. Moreover, AMA caused an altered pattern of expression of pluripotency markers (Sox2, OCT4) in mESCs. AMA further diminished mESC survival and proliferation and reduced the expression of cell proliferation marker, Ki-67. These changes coincided with altered expression of the epigenetic marker, Dnmt3a and other developmental regulators in a sex-dependent manner. Collectively, our data demonstrate the feasibility to utilise mESCs to reveal developmental mechanisms underlying AMA in the absence of maternal senescence and with reduced animal use.
Collapse
|
50
|
Al-Zubaidi U, Adhikari D, Cinar O, Zhang QH, Yuen WS, Murphy MP, Rombauts L, Robker RL, Carroll J. Mitochondria-targeted therapeutics, MitoQ and BGP-15, reverse aging-associated meiotic spindle defects in mouse and human oocytes. Hum Reprod 2021; 36:771-784. [PMID: 33367783 DOI: 10.1093/humrep/deaa300] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/28/2020] [Indexed: 12/22/2022] Open
Abstract
STUDY QUESTION Do mitochondria-targeted therapies reverse ageing- and oxidative stress-induced spindle defects in oocytes from mice and humans? SUMMARY ANSWER Exposure to MitoQ or BGP-15 during IVM protected against spindle and chromosomal defects in mouse oocytes exposed to oxidative stress or derived from reproductively aged mice whilst MitoQ promoted nuclear maturation and protected against chromosomal misalignments in human oocytes. WHAT IS KNOWN ALREADY Spindle and chromosomal abnormalities in oocytes are more prevalent with maternal aging, increasing the risk of aneuploidy, miscarriage and genetic disorders such as Down's syndrome. The origin of compromised oocyte function may be founded in mitochondrial dysfunction and increased reactive oxygen species (ROS). STUDY DESIGN, SIZE, DURATION Oocytes from young and old mice were treated with MitoQ and/or BGP-15 during IVM. To directly induce mitochondrial dysfunction, oocytes were treated with H2O2, and then treated the MitoQ and/or BGP-15. Immature human oocytes were cultured with or without MitoQ. Each experiment was repeated at least three times, and data were analyzed by unpaired-sample t-test or chi-square test. PARTICIPANTS/MATERIALS, SETTING, METHODS Immature germinal vesicle (GV) stage oocytes from 1-, 12- and 18-month-old mice were obtained from preovulatory ovarian follicles. Oocytes were treated with MitoQ and/or BGP-15 during IVM. GV-stage human oocytes were cultured with or without MitoQ. Mitochondrial membrane potential and mitochondrial ROS were measured by live-cell imaging. Meiotic spindle and chromosome alignments were visualized by immunofluorescent labeling of fixed oocytes and the 3-dimensional images were analyzed by Imaris. MAIN RESULTS AND THE ROLE OF CHANCE MitoQ or BGP-15 during IVM protects against spindle and chromosomal defects in oocytes exposed to oxidative stress and in oocytes from aged mice (P < 0.001). In human oocytes, the presence of MitoQ during IVM promoted nuclear maturation and had a similar positive effect in protecting against chromosomal misalignments (P < 0.001). LIMITATIONS, REASONS FOR CAUTION Our study identifies two excellent candidates that may help to improve fertility in older women. However, these potential therapies must be tested for efficacy in clinical IVM systems, and undergo thorough examination of resultant offspring in preclinical models before utilization. WIDER IMPLICATIONS OF THE FINDINGS Our results using in-vitro systems for oocyte maturation in both mouse and human provide proof of principle that mitochondrially targeted molecules such as MitoQ and BGP-15 may represent a novel therapeutic approach against maternal aging-related spindle and chromosomal abnormalities. STUDY FUNDING/COMPETING INTEREST(S) The project was financially supported by the National Health and Medical Research Council and Australian Research Council, Australia. U.A.-Z. was supported by the Iraqi Higher Education and Scientific Research Ministry PhD scholarship and O.C. was supported by TUBITAK-1059B191601275. M.P.M. consults for MitoQ Inc. and holds patents in mitochondria-targeted therapies. R.L.R. is an inventor on patents relating to the use of BGP-15 to improve gamete quality. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Usama Al-Zubaidi
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia.,Applied Embryology Department, High Institute for Infertility Diagnosis and Assisted Reproductive Technologies, AL-Nahrain University, Baghdad, Iraq
| | - Deepak Adhikari
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Ozgur Cinar
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia.,Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
| | - Qing-Hua Zhang
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Wai Shan Yuen
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Luk Rombauts
- Monash IVF, Melbourne, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Rebecca L Robker
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia.,School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA 5005, Australia
| | - John Carroll
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| |
Collapse
|