1
|
Li C, Li J, Du S, Ma Y, Guo Y, Zhang X, Wang B, Zhu S, An H, Chen M, Guo J, Han L, Ge J, Qian X, Schedl T, Guo X, Wang Q. FTDC1/2, oocyte-specific cofactors of DNMT1 required for epigenetic regulation and embryonic development. Cell Death Differ 2025:10.1038/s41418-025-01518-3. [PMID: 40295817 DOI: 10.1038/s41418-025-01518-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
The unique epigenetic patterns during gametogenesis and embryonic development indicate the existence of specialized methylation machinery. In the present study, we describe the discovery of two oocyte-specific cofactors of DNA methyltransferase 1 (DNMT1), encoded by uncharacterized genes, ferritin domain containing 1 and 2 (Ftdc1 and Ftdc2). Genetic ablation of Ftdc1 or Ftdc2 causes midgestation defects and female infertility. FTDC1 or FTDC2 depletion induces the progressive loss of DNA methylation including imprinted regions in early embryos. This loss correlates with a marked reduction in DNMT1 protein due to increased degradation, likely via the ubiquitin-proteasome pathway. Mechanistically, we find that FTDC1, FTDC2 and DNMT1 form a complex by direct interactions, thereby stabilizing each other. Surprisingly, knockout of Ftdc1 or Ftdc2 displayed stronger DNA demethylation phenotypes and earlier embryonic lethality than the Dnmt1-null mutant, implying their unique functions. These data suggest that FTDC1/2 are crucial players specifically involved in maintaining genomic methylation during embryogenesis, offering new insights into the epigenetic control of mammalian development.
Collapse
Affiliation(s)
- Congyang Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Jiashuo Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Siyu Du
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Yunfei Ma
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Xiangzheng Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Bing Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Huiqing An
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Ming Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Junjie Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Longsen Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Xu Qian
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Tim Schedl
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China.
| | - Qiang Wang
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China.
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Chen L, Wang WJ, Liu SY, Su RB, Wu YK, Wu X, Zhang SY, Qiao J, Sha QQ, Fan HY. NAT10-mediated mRNA N4-acetylation is essential for the translational regulation during oocyte meiotic maturation in mice. SCIENCE ADVANCES 2025; 11:eadp5163. [PMID: 39982985 PMCID: PMC11844725 DOI: 10.1126/sciadv.adp5163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 01/17/2025] [Indexed: 02/23/2025]
Abstract
The precise translational regulation of maternal messenger RNAs (mRNAs) drives mammalian oocyte maturation. However, the function and mechanism of posttranscriptional chemical modifications, especially the newly identified N4-acetylcytidine (ac4C) modification catalyzed by N-acetyltransferase 10 (NAT10), are unknown. In this study, we developed a low-input ac4C sequencing technology, ac4C LACE-seq, and mapped 8241 ac4C peaks at the whole-transcriptome level using 50 mouse oocytes at the germinal vesicle stage. Oocyte-specific Nat10 knockout wiped out ac4C signals in oocytes and caused severe defects in meiotic maturation and female infertility. Mechanically, Nat10 deletion led to a failure of ac4C deposition on mRNAs encoding key maternal factors, which regulate transcriptome stability and maternal-to-zygotic transition. Nat10-deleted oocytes showed decreased mRNA translation efficiency due to the direct inhibition of ac4C sites on specific transcripts during meiotic maturation. In summary, we developed a low-input, high-sensitivity mRNA ac4C profiling approach and highlighted the important physiological function of ac4C in the precise regulation of oocyte meiotic maturation by enhancing translation efficiency.
Collapse
Affiliation(s)
- Lu Chen
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Wen-Jing Wang
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Shao-Yuan Liu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Rui-Bao Su
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Yu-Ke Wu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Xuan Wu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Song-Ying Zhang
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Qian-Qian Sha
- College of Life Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Heng-Yu Fan
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Center for Biomedical Research, Shaoxing Institute, Zhejiang University, Shaoxing 312000, China
| |
Collapse
|
3
|
Wang H, Gao M, Cheng Q, Zhu S, Chen Y, Gu L, Guo X, Huo R, Xiong B, Wang Q. PAK4 promotes the cytoskeletal organization and meiotic maturation via phosphorylating DDX17 in oocyte. Cell Commun Signal 2025; 23:85. [PMID: 39948582 PMCID: PMC11827469 DOI: 10.1186/s12964-025-02085-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/05/2025] [Indexed: 02/16/2025] Open
Abstract
PAK4 has been widely reported to function in somatic cells. However, its role and the underlying mechanisms in meiotic oocytes are largely unknown. Here, we show that PAK4 deficiency significantly disrupts maturational progression and meiotic apparatus in mouse oocytes. Furthermore, based on the kinase substrate binding preference and systematic functional screening, our mechanistic investigation demonstrated that PAK4 promotes cytoskeletal organization and oocyte maturation through phosphorylating serine 597 on DDX17. Of note, we identified a marked reduction of PAK4 protein in oocytes from diabetic mice. Importantly, ectopic expression of hyperphosphorylation-mimicking DDX17 mutant (DDX17-S597D) partly prevented the meiotic defects in these diabetic oocytes, indicating that the decreased phosphorylation of DDX17 due to PAK4 insufficiency is responsible for the impaired oocyte quality. In sum, these findings unveil the pivotal role of PAK4 in oocyte development and indicate a novel mechanism controlling meiotic progression and structure.
Collapse
Affiliation(s)
- Hengjie Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Ming Gao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Qing Cheng
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Yu Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Ling Gu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Ran Huo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China.
| | - Bo Xiong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
- College of Animal Sciences, Zhejiang University, Hangzhou, China.
| | - Qiang Wang
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, 101 Longmian Rd, Nanjing, Jiangsu, 211166, China.
| |
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
|
Cheng S, Schuh M. Two mechanisms repress cyclin B1 translation to maintain prophase arrest in mouse oocytes. Nat Commun 2024; 15:10044. [PMID: 39567493 PMCID: PMC11579420 DOI: 10.1038/s41467-024-54161-w] [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: 01/18/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024] Open
Abstract
In mammals, oocytes are arrested in prophase of meiosis I for long periods of time. Prophase arrest is critical for reproduction because it allows oocytes to grow to their full size to support meiotic maturation and embryonic development. Prophase arrest requires the inhibitory phosphorylation of the mitotic kinase CDK1. Whether prophase arrest is also regulated at the translational level is unknown. Here, we show that prophase arrest is regulated by translational control of dormant cyclin B1 mRNAs. Using Trim-Away, we identify two mechanisms that maintain cyclin B1 dormancy and thus prophase arrest. First, a complex of the RNA-binding proteins DDX6, LSM14B and CPEB1 directly represses cyclin B1 translation through interacting with its 3'UTR. Second, cytoplasmic poly(A)-binding proteins (PABPCs) indirectly repress the translation of cyclin B1 and other poly(A)-tail-less or short-tailed mRNAs by sequestering the translation machinery on long-tailed mRNAs. Together, we demonstrate how RNA-binding proteins coordinately regulate prophase arrest, and reveal an unexpected role for PABPCs in controlling mRNA dormancy.
Collapse
Affiliation(s)
- Shiya Cheng
- Department of Meiosis, Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, TaiKang Center for Life and Medical Sciences, School of Basic Medical Sciences, Wuhan University, 430072, Wuhan, China
| | - Melina Schuh
- Department of Meiosis, Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37077, Göttingen, Germany.
| |
Collapse
|
6
|
Han Z, Wang R, Chi P, Zhang Z, Min L, Jiao H, Ou G, Zhou D, Qin D, Xu C, Gao Z, Qi Q, Li J, Lu Y, Wang X, Chen J, Yu X, Hu H, Li L, Deng D. The subcortical maternal complex modulates the cell cycle during early mammalian embryogenesis via 14-3-3. Nat Commun 2024; 15:8887. [PMID: 39406751 PMCID: PMC11480350 DOI: 10.1038/s41467-024-53277-3] [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: 02/25/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
The subcortical maternal complex (SCMC) is essential for safeguarding female fertility in mammals. Assembled in oocytes, the SCMC maintains the cleavage of early embryos, but the underlying mechanism remains unclear. Here, we report that 14-3-3, a multifunctional protein, is a component of the SCMC. By resolving the structure of the 14-3-3-containing SCMC, we discover that phosphorylation of TLE6 contributes to the recruitment of 14-3-3. Mechanistically, during maternal-to-embryo transition, the SCMC stabilizes 14-3-3 protein and contributes to the proper control of CDC25B, thus ensuring the activation of the maturation-promoting factor and mitotic entry in mouse zygotes. Notably, the SCMC establishes a conserved molecular link with 14-3-3 and CDC25B in human oocytes/embryos. This study discloses the molecular mechanism through which the SCMC regulates the cell cycle in early embryos and elucidates the function of the SCMC in mammalian early embryogenesis.
Collapse
Affiliation(s)
- Zhuo Han
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rui Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
- Department of Reproductive Medicine, the First People's Hospital of Yunnan Province, Kunming, China
| | - Pengliang Chi
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zihan Zhang
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ling Min
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Haizhan Jiao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| | - Guojin Ou
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- Clinical laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dan Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
| | - Dandan Qin
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
| | - Chengpeng Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
| | - Zheng Gao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qianqian Qi
- Clinical laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jialu Li
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yuechao Lu
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiang Wang
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- NHC key Laboratory of Chronobiology, Sichuan University, Chengdu, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Jing Chen
- Laboratory of Pediatric Surgery, Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xingjiang Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China.
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China.
| | - Dong Deng
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
- NHC key Laboratory of Chronobiology, Sichuan University, Chengdu, China.
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Sun H, Han L, Guo Y, An H, Wang B, Zhang X, Li J, Jiang Y, Wang Y, Sun G, Zhu S, Tang S, Ge J, Chen M, Guo X, Wang Q. The global phosphorylation landscape of mouse oocytes during meiotic maturation. EMBO J 2024; 43:4752-4785. [PMID: 39256562 PMCID: PMC11480333 DOI: 10.1038/s44318-024-00222-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
Phosphorylation is a key post-translational modification regulating protein function and biological outcomes. However, the phosphorylation dynamics orchestrating mammalian oocyte development remains poorly understood. In the present study, we apply high-resolution mass spectrometry-based phosphoproteomics to obtain the first global in vivo quantification of mouse oocyte phosphorylation. Of more than 8000 phosphosites, 75% significantly oscillate and 64% exhibit marked upregulation during meiotic maturation, indicative of the dominant regulatory role. Moreover, we identify numerous novel phosphosites on oocyte proteins and a few highly conserved phosphosites in oocytes from different species. Through functional perturbations, we demonstrate that phosphorylation status of specific sites participates in modulating critical events including metabolism, translation, and RNA processing during meiosis. Finally, we combine inhibitor screening and enzyme-substrate network prediction to discover previously unexplored kinases and phosphatases that are essential for oocyte maturation. In sum, our data define landscape of the oocyte phosphoproteome, enabling in-depth mechanistic insights into developmental control of germ cells.
Collapse
Affiliation(s)
- Hongzheng Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Longsen Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Huiqing An
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Bing Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Xiangzheng Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Jiashuo Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Yingtong Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Yue Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Guangyi Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Shoubin Tang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Minjian Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China.
- Department of Histology and Embryology, Nanjing Medical University, 211166, Nanjing, China.
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, China.
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China.
| |
Collapse
|
8
|
Tang F, Hummitzsch K, Rodgers RJ. RNAseq analysis of oocyte maturation from the germinal vesicle stage to metaphase II in pig and human. PLoS One 2024; 19:e0305893. [PMID: 39121087 PMCID: PMC11315340 DOI: 10.1371/journal.pone.0305893] [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: 03/17/2024] [Accepted: 06/06/2024] [Indexed: 08/11/2024] Open
Abstract
During maturation oocytes at the germinal vesicle (GV) stage progress to metaphase II (MII). However, during in vitro maturation a proportion often fail to progress. To understand these processes, we employed RNA sequencing to examine the transcriptome profile of these three groups of oocytes from the pig. We compared our findings with similar public oocyte data from humans. The transcriptomes in oocytes that failed to progress was similar to those that did. We found in both species, the most upregulated genes in MII oocytes were associated with chromosome segregation and cell cycle processes, while the most down regulated genes were relevant to ribosomal and mitochondrial pathways. Moreover, those genes involved in chromosome segregation during GV to MII transition were conserved in pig and human. We also compared MII and GV oocyte transcriptomes at the isoform transcript level in both species. Several thousands of genes (including DTNBP1, MAPK1, RAB35, GOLGA7, ATP1A1 and ATP2B1) identified as not different in expression at a gene transcript level were found to have differences in isoform transcript levels. Many of these genes were involved in ATPase-dependent or GTPase-dependent intracellular transport in pig and human, respectively. In conclusion, our study suggests the failure to progress to MII in vitro may not be regulated at the level of the genome and that many genes are differentially regulated at the isoform level, particular those involved ATPase- or GTPase-dependent intracellular transport.
Collapse
Affiliation(s)
- Feng Tang
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Katja Hummitzsch
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Raymond J. Rodgers
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
9
|
Chang H, Huang C, Cheng S, Li J, Wang X. Fbxo28 is essential for spindle migration and morphology during mouse oocyte meiosis I. Int J Biol Macromol 2024; 275:133232. [PMID: 38960234 DOI: 10.1016/j.ijbiomac.2024.133232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/28/2024] [Accepted: 06/15/2024] [Indexed: 07/05/2024]
Abstract
Spindle migration and assembly regulates asymmetric oocyte division, which is essential for fertility. Fbxo28, as a member of SCF (Skp1-Cul1-F-box) ubiquitin E3 ligases complex, is specifically expressed in oocytes. However, little is known about the functions of Fbxo28 in spindle assembly and migration during oocyte meiosis I. In present study, microinjection with morpholino oligonucleotides and exogenous mRNA for knockdown and rescue experiments, and immunofluorescence staining, western blot, timelapse confocal microscopy and chromosome spreading were utilized to explore the roles of Fbxo28 in asymmetric division during meiotic maturation. Our data suggested that Fbxo28 mainly localized at chromosomes and acentriolar microtubule-organizing centers (aMTOCs). Depletion of Fbxo28 did not affect polar body extrusion but caused defects in spindle morphology and migration, indicative of the failure of asymmetric division. Moreover, absence of Fbxo28 disrupted both cortical and cytoplasmic actin assembly and decreased the expression of ARPC2 and ARP3. These defects could be rescued by exogenous Fbxo28-myc mRNA supplement. Collectively, this study demonstrated that Fbxo28 affects spindle morphology and actin-based spindle migration during mouse oocyte meiotic maturation.
Collapse
Affiliation(s)
- Haoya Chang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China; Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Chenyang Huang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Siyu Cheng
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jian Li
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Xiaohong Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China.
| |
Collapse
|
10
|
Jin Y, Sun G, Li J, Cheng Q, Sun H, Han L, Guo X, Zhu S, Wang Q. MIB2 Functions in Oocyte Meiosis by Modulating Chromatin Configuration. Mol Cell Proteomics 2024; 23:100813. [PMID: 39019259 PMCID: PMC11364126 DOI: 10.1016/j.mcpro.2024.100813] [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: 01/10/2024] [Revised: 06/13/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024] Open
Abstract
Chromatin configuration serves as a principal indicator of GV (germinal vesicle)-stage oocyte quality. However, the underlying mechanisms governing the chromatin configuration transition from NSN (non-surrounded nucleolus) to SN (surrounded nucleolus) remain unclear. In this study, by conducting a quantitative proteomic analysis, we identified an increased expression of the MIB2 (MIB E3 ubiquitin protein ligase 2) protein in SN oocytes. Specific depletion of MIB2 in SN oocytes not only leads to severe disruption of the meiotic apparatus and a higher incidence of aneuploidy but also adversely affects meiotic maturation and early embryo development. Notably, overexpression of MIB2 in NSN oocytes facilitates the chromatin configuration transition. Meantime, we observed that forced expression of MIB2 in NSN oocytes significantly mitigates spindle/chromosome disorganization and aneuploidy. In summary, our results suggest that chromatin configuration transition regulated by MIB2 is crucial for oocytes to acquire developmental competence.
Collapse
Affiliation(s)
- Yifei Jin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Guangyi Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Jiashuo Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Qing Cheng
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Hongzheng Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Longsen Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China.
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
11
|
Fulka H. ELVAs: The new 'super-organelles' of the oocyte. Lab Anim (NY) 2024; 53:133-134. [PMID: 38783098 DOI: 10.1038/s41684-024-01379-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Affiliation(s)
- Helena Fulka
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
12
|
Jacques F, Tichopád T, Demko M, Bystrý V, Křížová KC, Seifertová M, Voříšková K, Fuad MMH, Vetešník L, Šimková A. Reproduction-associated pathways in females of gibel carp (Carassius gibelio) shed light on the molecular mechanisms of the coexistence of asexual and sexual reproduction. BMC Genomics 2024; 25:548. [PMID: 38824502 PMCID: PMC11144346 DOI: 10.1186/s12864-024-10462-4] [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: 05/28/2024] [Indexed: 06/03/2024] Open
Abstract
Gibel carp (Carassius gibelio) is a cyprinid fish that originated in eastern Eurasia and is considered as invasive in European freshwater ecosystems. The populations of gibel carp in Europe are mostly composed of asexually reproducing triploid females (i.e., reproducing by gynogenesis) and sexually reproducing diploid females and males. Although some cases of coexisting sexual and asexual reproductive forms are known in vertebrates, the molecular mechanisms maintaining such coexistence are still in question. Both reproduction modes are supposed to exhibit evolutionary and ecological advantages and disadvantages. To better understand the coexistence of these two reproduction strategies, we performed transcriptome profile analysis of gonad tissues (ovaries) and studied the differentially expressed reproduction-associated genes in sexual and asexual females. We used high-throughput RNA sequencing to generate transcriptomic profiles of gonadal tissues of triploid asexual females and males, diploid sexual males and females of gibel carp, as well as diploid individuals from two closely-related species, C. auratus and Cyprinus carpio. Using SNP clustering, we showed the close similarity of C. gibelio and C. auratus with a basal position of C. carpio to both Carassius species. Using transcriptome profile analyses, we showed that many genes and pathways are involved in both gynogenetic and sexual reproduction in C. gibelio; however, we also found that 1500 genes, including 100 genes involved in cell cycle control, meiosis, oogenesis, embryogenesis, fertilization, steroid hormone signaling, and biosynthesis were differently expressed in the ovaries of asexual and sexual females. We suggest that the overall downregulation of reproduction-associated pathways in asexual females, and their maintenance in sexual ones, allows the populations of C. gibelio to combine the evolutionary and ecological advantages of the two reproductive strategies. However, we showed that many sexual-reproduction-related genes are maintained and expressed in asexual females, suggesting that gynogenetic gibel carp retains the genetic toolkits for meiosis and sexual reproduction. These findings shed new light on the evolution of this asexual and sexual complex.
Collapse
Affiliation(s)
- Florian Jacques
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 611 37, Czech Republic.
| | - Tomáš Tichopád
- Laboratory of Non-Mendelian Evolution, Institute of Animal Physiology and Genetics of the CAS, Liběchov, 277 21, Czech Republic
- Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, Vodňany, 389 25, Czech Republic
| | - Martin Demko
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 611 37, Czech Republic
- Central European Institute of Technology, Masaryk University, Brno, 625 00, Czech Republic
| | - Vojtěch Bystrý
- Central European Institute of Technology, Masaryk University, Brno, 625 00, Czech Republic
| | - Kristína Civáňová Křížová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 611 37, Czech Republic
| | - Mária Seifertová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 611 37, Czech Republic
| | - Kristýna Voříšková
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 611 37, Czech Republic
| | - Md Mehedi Hasan Fuad
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 611 37, Czech Republic
| | - Lukáš Vetešník
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 611 37, Czech Republic
- Institute of Vertebrate Biology, Czech Academy of Science, Květná 8, Brno, 603 65, Czech Republic
| | - Andrea Šimková
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 611 37, Czech Republic
| |
Collapse
|
13
|
Sachs MK, Makieva S, Velasco Gil A, Xie M, Ille F, Salvadori V, Schmidhauser M, Saenz-de-Juano MD, Ulbrich SE, Leeners B. Transcriptomic signature of luteinized cumulus cells of oocytes developing to live birth after women received intracytoplasmic sperm injection. F&S SCIENCE 2024; 5:24-38. [PMID: 38036000 DOI: 10.1016/j.xfss.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023]
Abstract
OBJECTIVE To compare the transcriptome of human cumulus cells (CCs) from oocytes with different outcomes (pregnancy yes/no, live birth [LB] yes/no), to identify noninvasive biomarkers for oocyte selection as well as new therapeutic targets to increase LB rates from assisted reproductive technologies (ART). DESIGN Retrospective observational study. SETTINGS This study was conducted at a University Hospital in Switzerland. PATIENTS Subfertile couples undergoing controlled ovarian superstimulation and intracytoplasmic sperm injection with subsequent unbiopsied embryo transfer below the female age of 43 years. INTERVENTION(S) RNA sequencing of CCs from oocytes results in a pregnancy, no pregnancy, LB, or no LB. MAIN OUTCOME MEASURES Differential gene expression (DEG) between CCs of oocytes results in "no pregnancy" vs. "pregnancy" and "pregnancy only" vs. "live birth." RESULTS Although RNA sequencing did not reveal DEGs when comparing the transcriptomic profiles of the groups "no pregnancy" with "pregnancy," we identified 139 DEGs by comparing "pregnancy only" with "live birth," of which 28 belonged to clusters relevant to successful ART outcomes (i.e., CTGF, SERPINE2, PCK1, HHIP, HS3ST, and BIRC5). A functional enrichment analysis revealed that the transcriptome of CCs associated with LB depicts pathways of extracellular matrix, inflammatory cascades leading to ovulation, cell patterning, proliferation, and differentiation, and silencing pathways leading to apoptosis. CONCLUSION We identified a CCs transcriptomic profile associated with LB after embryo transfer that, after further validation, could serve to predict successful ART outcomes. The definition of relevant pathways of CCs related to oocyte competency contributes to a broader understanding of the cumulus oocyte complex and helps identify further therapeutic targets for improving ART success.
Collapse
Affiliation(s)
- Maike K Sachs
- Department of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse, Zurich, Switzerland.
| | - Sofia Makieva
- Department of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse, Zurich, Switzerland
| | - Ana Velasco Gil
- Department of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse, Zurich, Switzerland
| | - Min Xie
- Department of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse, Zurich, Switzerland
| | - Fabian Ille
- Center of Bioscience and Medical Engineering, Institute of Medical Engineering, Lucerne University of Applied Sciences and Arts, Obermattweg, Hergiswil, Switzerland
| | - Vincent Salvadori
- Center of Bioscience and Medical Engineering, Institute of Medical Engineering, Lucerne University of Applied Sciences and Arts, Obermattweg, Hergiswil, Switzerland
| | - Meret Schmidhauser
- ETH Zürich, Animal Physiology, Institute of Agricultural Sciences, Universitätsstrasse Zurich, Switzerland
| | - Mara D Saenz-de-Juano
- ETH Zürich, Animal Physiology, Institute of Agricultural Sciences, Universitätsstrasse Zurich, Switzerland
| | - Susanne E Ulbrich
- ETH Zürich, Animal Physiology, Institute of Agricultural Sciences, Universitätsstrasse Zurich, Switzerland
| | - Brigitte Leeners
- Department of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse, Zurich, Switzerland
| |
Collapse
|
14
|
Wu X, Wang S, Guo Y, Song S, Zeng S. KAT8 functions in redox homeostasis and mitochondrial dynamics during mouse oocyte meiosis progression. FASEB J 2024; 38:e23435. [PMID: 38243686 DOI: 10.1096/fj.202301946r] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 01/21/2024]
Abstract
As a histone acetyltransferase, lysine acetyltransferase 8 (KAT8) participates in diverse biological processes. However, the effect of KAT8 on oocyte maturation in mice remains unclear. In this study, we found that mouse oocytes overexpressing Kat8-OE induced maturation failure manifested reduced rates of GVBD and first polar body emission. In addition, immunostaining results revealed that Kat8 overexpressing oocytes showed inappropriate mitochondrial distribution patterns, overproduction of reactive oxygen species (ROS), accumulation of phosphorylated γH2AX, hyperacetylation of α-tubulin, and severely disrupted spindle/chromosome organization. Moreover, we revealed that Kat8 overexpression induced a decline in SOD1 proteins and KAT8's interaction with SOD1 in mouse ovaries via immunoprecipitation. Western blotting data confirmed that Kat8-OE induced downregulation of SOD1 expression, which is a key factor for the decline of oocyte quality in advanced maternal age. Also, the injection of Myc-Sod1 cRNA could partially rescue maternal age-induced meiotic defects in oocytes. In conclusion, our data demonstrated that high level of KAT8 inhibited SOD1 activity, which in turn induced defects of mitochondrial dynamics, imbalance of redox homeostasis, and spindle/chromosome disorganization during mouse oocyte maturation.
Collapse
Affiliation(s)
- Xuan Wu
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shiwei Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yajun Guo
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuang Song
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shenming Zeng
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
15
|
Yu T, Zhang C, Song W, Zhao X, Cheng Y, Liu J, Su J. Single-cell RNA-seq and single-cell bisulfite-sequencing reveal insights into yak preimplantation embryogenesis. J Biol Chem 2024; 300:105562. [PMID: 38097189 PMCID: PMC10821408 DOI: 10.1016/j.jbc.2023.105562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 11/17/2023] [Accepted: 12/03/2023] [Indexed: 01/13/2024] Open
Abstract
Extensive epigenetic reprogramming occurs during preimplantation embryonic development. However, the impact of DNA methylation in plateau yak preimplantation embryos and how epigenetic reprogramming contributes to transcriptional regulatory networks are unclear. In this study, we quantified gene expression and DNA methylation in oocytes and a series of yak embryos at different developmental stages and at single-cell resolution using single-cell bisulfite-sequencing and RNA-seq. We characterized embryonic genome activation and maternal transcript degradation and mapped epigenetic reprogramming events critical for embryonic development. Through cross-species transcriptome analysis, we identified 31 conserved maternal hub genes and 39 conserved zygotic hub genes, including SIN3A, PRC1, HDAC1/2, and HSPD1. Notably, by combining single-cell DNA methylation and transcriptome analysis, we identified 43 candidate methylation driver genes, such as AURKA, NUSAP1, CENPF, and PLK1, that may be associated with embryonic development. Finally, using functional approaches, we further determined that the epigenetic modifications associated with the histone deacetylases HDAC1/2 are essential for embryonic development and that the deubiquitinating enzyme USP7 may affect embryonic development by regulating DNA methylation. Our data represent an extensive resource on the transcriptional dynamics of yak embryonic development and DNA methylation remodeling, and provide new insights into strategies for the conservation of germplasm resources, as well as a better understanding of mammalian early embryonic development that can be applied to investigate the causes of early developmental disorders.
Collapse
Affiliation(s)
- Tong Yu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Chengtu Zhang
- Academician Zhang Yong Innovation Center, Xining Animal Disease Control Center, Xining, Qinghai, China
| | - Weijia Song
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xinyi Zhao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuyao Cheng
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jun Liu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Jianmin Su
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
16
|
Chen M, Yang W, Guo Y, Hou X, Zhu S, Sun H, Guo X, Chen M, Wang Q. Multi-omics reveal the metabolic patterns in mouse cumulus cells during oocyte maturation. J Ovarian Res 2023; 16:156. [PMID: 37550748 PMCID: PMC10408154 DOI: 10.1186/s13048-023-01237-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/13/2023] [Indexed: 08/09/2023] Open
Abstract
Bi-directional communication between cumulus cells and the surrounded oocytes is important for the development and functions of both compartments. However, the metabolic framework in cumulus cells has not been systematically described. In the present study, cumulus cells from cumulus-oocyte complexes (COCs) at three key time points were isolated (arrested GV stage, post-hCG 0h; meiotic resumption GVBD stage, post-hCG 3h; and metaphase II stage, post-hCG 12h), and the temporal metabolomic and proteomic profiling were performed. Integrated multi-omics analysis reveals the global metabolic patterns in cumulus cells during mouse oocyte maturation. In particular, we found the active hyaluronic acid metabolism, steroid hormone synthesis, and prostaglandin E2 (PGE2) production in cumulus cells. Meanwhile, accompanying the oocyte maturation, a progressive increase in nucleotide and amino acid metabolism was detected in the surrounding cumulus cells. In sum, the data serve as a valuable resource for probing metabolism during terminal differentiation of ovarian granulosa cells, and provide the potential biomarkers for improving and predicting oocyte quality.
Collapse
Affiliation(s)
- Ming Chen
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, 101 Longmian Rd, Nanjing, Jiangsu, 211166, China
| | - Weizheng Yang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, 101 Longmian Rd, Nanjing, Jiangsu, 211166, China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, 101 Longmian Rd, Nanjing, Jiangsu, 211166, China
| | - Xiaojing Hou
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, 101 Longmian Rd, Nanjing, Jiangsu, 211166, China
| | - Hongzheng Sun
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, 101 Longmian Rd, Nanjing, Jiangsu, 211166, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, 101 Longmian Rd, Nanjing, Jiangsu, 211166, China.
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Minjian Chen
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, 101 Longmian Rd, Nanjing, Jiangsu, 211166, China.
- Department of Histology and Embryology, Nanjing Medical University, Nanjing, 211166, China.
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, 101 Longmian Rd, Nanjing, Jiangsu, 211166, China.
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
17
|
Ma X, Wang M, Wang J, Zhang Q, Pu S, Wang R, Yu S, Wang L, Pan Y. Dynamic Changes in Proteome during Yak Oocyte Maturation Analyzed Using iTRAQ Technology. Animals (Basel) 2023; 13:2085. [PMID: 37443883 DOI: 10.3390/ani13132085] [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: 04/28/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of this study was to investigate protein regulation at different time points during the in vitro maturation of yak oocytes. Yak oocytes at GV, MI, and MII stages were collected during in vitro maturation, and differential proteomics sequencing was performed using iTRAQ technology. GO functional classification indicated that the differential proteins were closely associated with biological processes such as "metabolic processes", and molecular events such as "binding" molecular-function-related categories were active. KOG analysis showed that energy-metabolism-related activities were vigorous during oocyte development from the GV phase to MI phase, and genetic material preparation activities were more active when oocytes developed from the MI stage to MII stage. KEGG pathway analysis showed that the PPAR metabolic pathway, Hippo signaling pathway, and ECM-receptor interaction and metabolic pathway were enriched from the GV to the MI stages. The PI3K-Akt, TGF-β, and phagosome pathways were enriched from the MI stage to the MII stage. These results indicate that transient dynamic changes occurred in the proteome during the maturation of yak oocytes, and the physiological functions mediated by these were also different. The accurate identification of the differential proteins in the three stages of GV, MI, and MII was helpful in further analyzing the molecular regulatory mechanism of yak oocyte maturation.
Collapse
Affiliation(s)
- Xin Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou 730070, China
| | - Meng Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou 730070, China
| | - Jinglei Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou 730070, China
| | - Qian Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou 730070, China
| | - Sisi Pu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Rui Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Sijiu Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou 730070, China
| | - Libin Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou 730070, China
| | - Yangyang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou 730070, China
| |
Collapse
|