1
|
Xu X, Jiang H, Wang D, Rehman SU, Li Z, Song X, Cui K, Luo X, Yang C, Liu Q. Exploration of transcriptional regulation network between buffalo oocytes and granulosa cells and its impact on different diameter follicles. BMC Genomics 2024; 25:1004. [PMID: 39462339 PMCID: PMC11515274 DOI: 10.1186/s12864-024-10912-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Buffalo is a globally important livestock species, but its reproductive performance is relatively low than cattles. At present, dominant follicle development specific process and mechanistic role of follicular growth related genes in water buffaloes are not well understood. Therefore, we comprehensively performed transcriptomics of granulosa cells and oocytes from different-sized follicles in water buffalo to identify key candidate genes that influence follicle development and diameter, and further explored the potential regulatory mechanisms of granulosa cells and oocytes in the process of water buffalo follicle development. RESULTS In this study, we found918 granulosa cell transcripts and 1401 oocyte transcripts were correlated in follicles of different diameters, and the expression differences were significant. Subsequent enrichment analysis of the co-expressed differentially expressed transcripts identified several genes targeted by long non-coding RNAs (lncRNAs) and associated with follicular development. Notably, the upregulation of BUB1 regulated by MSTRG.41325.4 and interactive action of SMAD2 and SMAD7 might have key regulatory role in follicular development. Additionally, we also detected key differentially expressed genes that potentially influence follicular hormone metabolism and growth, like ID2, CHRD, TGIF2 and MAD2L1, and constructed an interaction network between lncRNA transcripts and mRNAs. CONCLUSIONS In summary, this study preliminarily revealed the differences in gene expression patterns among buffalo follicles of different sizes and their potential molecular regulatory mechanisms. It provides a new perspective for exploring the mechanism of buffalo follicular dominance and improving buffalo reproductive performance.
Collapse
Affiliation(s)
- Xiaoxian Xu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Hancai Jiang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Dong Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Saif Ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Zhipeng Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Xinhui Song
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Kuiqing Cui
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Xier Luo
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Chunyan Yang
- Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning, 530001, China.
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China.
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China.
| |
Collapse
|
2
|
Sciorio R, Campos G, Tramontano L, Bulletti FM, Baldini GM, Vinciguerra M. Exploring the effect of cryopreservation in assisted reproductive technology and potential epigenetic risk. ZYGOTE 2023; 31:420-432. [PMID: 37409505 DOI: 10.1017/s0967199423000345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Since the birth of the first baby by in vitro fertilization in 1978, more than 9 million children have been born worldwide using medically assisted reproductive treatments. Fertilization naturally takes place in the maternal oviduct where unique physiological conditions enable the early healthy development of the embryo. During this dynamic period of early development major waves of epigenetic reprogramming, crucial for the normal fate of the embryo, take place. Increasingly, over the past 20 years concerns relating to the increased incidence of epigenetic anomalies in general, and genomic-imprinting disorders in particular, have been raised following assisted reproduction technology (ART) treatments. Epigenetic reprogramming is particularly susceptible to environmental conditions during the periconceptional period and non-physiological conditions such as ovarian stimulation, in vitro fertilization and embryo culture, as well as cryopreservation procedure, might have the potential to independently or collectively contribute to epigenetic dysregulation. Therefore, this narrative review offers a critical reappraisal of the evidence relating to the association between embryo cryopreservation and potential epigenetic regulation and the consequences on gene expression together with long-term consequences for offspring health and wellbeing. Current literature suggests that epigenetic and transcriptomic profiles are sensitive to the stress induced by vitrification, in terms of osmotic shock, temperature and pH changes, and toxicity of cryoprotectants, it is therefore, critical to have a more comprehensive understanding and recognition of potential unanticipated iatrogenic-induced perturbations of epigenetic modifications that may or may not be a consequence of vitrification.
Collapse
Affiliation(s)
- Romualdo Sciorio
- Edinburgh Assisted Conception Programme, EFREC, Royal Infirmary of Edinburgh, UK
| | | | - Luca Tramontano
- Department of Women, Infants and Adolescents, Division of Obstetrics, Geneve University Hospitals, Boulevard de la Cluse 30, Geneve 14, Switzerland
| | - Francesco M Bulletti
- Department Obstetrics and Gynecology, University Hospital of Vaud, Lausanne, Switzerland
| | | | - Marina Vinciguerra
- Department of Biomedical Sciences and Human Oncology, Obstetrics and Gynaecology Section, University of Bari, Italy
- Clinic of Obstetrics and Gynecology 'Santa Caterina Novella', Galatina Hospital, Italy
| |
Collapse
|
3
|
Camlin NJ, Venkatachalam I, Evans JP. Oscillations in PP1 activity are essential for accurate progression through mammalian oocyte meiosis. Cell Cycle 2023; 22:1614-1636. [PMID: 37340734 PMCID: PMC10361142 DOI: 10.1080/15384101.2023.2225924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/17/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
Tightly controlled fluctuations in kinase and phosphatase activity play important roles in regulating M-phase transitions. Protein Phosphatase 1 (PP1) is one of these phosphatases, with oscillations in PP1 activity driving mitotic M-phase. Evidence from a variety of experimental systems also points to roles in meiosis. Here, we report that PP1 is important for M-phase transitions through mouse oocyte meiosis. We employed a unique small-molecule approach to inhibit or activate PP1 at distinct phases of mouse oocyte meiosis. These studies show that temporal control of PP1 activity is essential for the G2/M transition, metaphase I/anaphase I transition, and the formation of a normal metaphase II oocyte. Our data also reveal that inappropriate activation of PP1 is more deleterious at the G2/M transition than at prometaphase I-to-metaphase I, and that an active pool of PP1 during prometaphase is vital for metaphase I/anaphase I transition and metaphase II chromosome alignment. Taken together, these results establish that loss of oscillations in PP1 activity causes a range of severe meiotic defects, pointing to essential roles for PP1 in female fertility, and more broadly, M-phase regulation.
Collapse
Affiliation(s)
- Nicole J. Camlin
- Department of Biological Sciences, Purdue University, West Lafayette, INUnited States
| | - Ilakkiya Venkatachalam
- Department of Biological Sciences, Purdue University, West Lafayette, INUnited States
- Department of Human Genetics, University of Michigan, Ann Arbor, MIUnited States
| | - Janice P. Evans
- Department of Biological Sciences, Purdue University, West Lafayette, INUnited States
| |
Collapse
|
4
|
Wang J, Sun X, Yang Z, Li S, Wang Y, Ren R, Liu Z, Yu D. Epigenetic regulation in premature ovarian failure: A literature review. Front Physiol 2023; 13:998424. [PMID: 36685174 PMCID: PMC9846267 DOI: 10.3389/fphys.2022.998424] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/14/2022] [Indexed: 01/05/2023] Open
Abstract
Premature ovarian failure (POF), or premature ovarian insufficiency (POI), is a multifactorial and heterogeneous disease characterized by amenorrhea, decreased estrogen levels and increased female gonadotropin levels. The incidence of POF is increasing annually, and POF has become one of the main causes of infertility in women of childbearing age. The etiology and pathogenesis of POF are complex and have not yet been clearly elucidated. In addition to genetic factors, an increasing number of studies have revealed that epigenetic changes play an important role in the occurrence and development of POF. However, we found that very few papers have summarized epigenetic variations in POF, and a systematic analysis of this topic is therefore necessary. In this article, by reviewing and analyzing the most relevant literature in this research field, we expound on the relationship between DNA methylation, histone modification and non-coding RNA expression and the development of POF. We also analyzed how environmental factors affect POF through epigenetic modulation. Additionally, we discuss potential epigenetic biomarkers and epigenetic treatment targets for POF. We anticipate that our paper may provide new therapeutic clues for improving ovarian function and maintaining fertility in POF patients.
Collapse
Affiliation(s)
- Jing Wang
- Department of Reproductive Medicine, Department of Prenatal Diagnosis, Changchun, China
| | | | | | - Sijie Li
- Department of Breast Surgery, Changchun, China
| | - Yufeng Wang
- Public Research Platform, The First Hospital of Jilin University, Jilin, China
| | - Ruoxue Ren
- Public Research Platform, The First Hospital of Jilin University, Jilin, China
| | - Ziyue Liu
- Public Research Platform, The First Hospital of Jilin University, Jilin, China
| | - Dehai Yu
- Public Research Platform, The First Hospital of Jilin University, Jilin, China,*Correspondence: Dehai Yu,
| |
Collapse
|
5
|
Zhao S, Wang H, Hu Z, Sahlu BW, Heng N, Gong J, Wang H, Zhu H. Identification of spermatogenesis-related lncRNA in Holstein bull testis after sexual maturity based on transcriptome analysis. Anim Reprod Sci 2022; 247:107146. [DOI: 10.1016/j.anireprosci.2022.107146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/29/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
|
6
|
Wu Y, Li M, Yang M. Post-Translational Modifications in Oocyte Maturation and Embryo Development. Front Cell Dev Biol 2021; 9:645318. [PMID: 34150752 PMCID: PMC8206635 DOI: 10.3389/fcell.2021.645318] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
Mammalian oocyte maturation and embryo development are unique biological processes regulated by various modifications. Since de novo mRNA transcription is absent during oocyte meiosis, protein-level regulation, especially post-translational modification (PTM), is crucial. It is known that PTM plays key roles in diverse cellular events such as DNA damage response, chromosome condensation, and cytoskeletal organization during oocyte maturation and embryo development. However, most previous reviews on PTM in oocytes and embryos have only focused on studies of Xenopus laevis or Caenorhabditis elegans eggs. In this review, we will discuss the latest discoveries regarding PTM in mammalian oocytes maturation and embryo development, focusing on phosphorylation, ubiquitination, SUMOylation and Poly(ADP-ribosyl)ation (PARylation). Phosphorylation functions in chromosome condensation and spindle alignment by regulating histone H3, mitogen-activated protein kinases, and some other pathways during mammalian oocyte maturation. Ubiquitination is a three-step enzymatic cascade that facilitates the degradation of proteins, and numerous E3 ubiquitin ligases are involved in modifying substrates and thus regulating oocyte maturation, oocyte-sperm binding, and early embryo development. Through the reversible addition and removal of SUMO (small ubiquitin-related modifier) on lysine residues, SUMOylation affects the cell cycle and DNA damage response in oocytes. As an emerging PTM, PARlation has been shown to not only participate in DNA damage repair, but also mediate asymmetric division of oocyte meiosis. Each of these PTMs and external environments is versatile and contributes to distinct phases during oocyte maturation and embryo development.
Collapse
Affiliation(s)
- Yu Wu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Mo Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Mo Yang
- Medical Center for Human Reproduction, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
He M, Zhang T, Yang Y, Wang C. Mechanisms of Oocyte Maturation and Related Epigenetic Regulation. Front Cell Dev Biol 2021; 9:654028. [PMID: 33842483 PMCID: PMC8025927 DOI: 10.3389/fcell.2021.654028] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Meiosis is the basis of sexual reproduction. In female mammals, meiosis of oocytes starts before birth and sustains at the dictyate stage of meiotic prophase I before gonadotropins-induced ovulation happens. Once meiosis gets started, the oocytes undergo the leptotene, zygotene, and pachytene stages, and then arrest at the dictyate stage. During each estrus cycle in mammals, or menstrual cycle in humans, a small portion of oocytes within preovulatory follicles may resume meiosis. It is crucial for females to supply high quality mature oocytes for sustaining fertility, which is generally achieved by fine-tuning oocyte meiotic arrest and resumption progression. Anything that disturbs the process may result in failure of oogenesis and seriously affect both the fertility and the health of females. Therefore, uncovering the regulatory network of oocyte meiosis progression illuminates not only how the foundations of mammalian reproduction are laid, but how mis-regulation of these steps result in infertility. In order to provide an overview of the recently uncovered cellular and molecular mechanism during oocyte maturation, especially epigenetic modification, the progress of the regulatory network of oocyte meiosis progression including meiosis arrest and meiosis resumption induced by gonadotropins is summarized. Then, advances in the epigenetic aspects, such as histone acetylation, phosphorylation, methylation, glycosylation, ubiquitination, and SUMOylation related to the quality of oocyte maturation are reviewed.
Collapse
Affiliation(s)
- Meina He
- Department of Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Tuo Zhang
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Yi Yang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan, China
| |
Collapse
|
8
|
Wang YY, Duan SH, Wang GL, Li JL. Integrated mRNA and miRNA expression profile analysis of female and male gonads in Hyriopsis cumingii. Sci Rep 2021; 11:665. [PMID: 33436779 PMCID: PMC7804246 DOI: 10.1038/s41598-020-80264-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/18/2020] [Indexed: 01/29/2023] Open
Abstract
Hyriopsis cumingii is an important species for freshwater pearl cultivation in China. In terms of pearl production, males have larger pearls and better glossiness than females, but there are few reports focusing on the sex of H. cumingii. In this study, six mRNA and six microRNA (miRNA) libraries were prepared from ovaries and testes. Additionally, 28,502 differentially expressed genes (DEGs) and 32 differentially expressed miRNAs (DEMs) were identified. Compared with testis, 14,360 mRNAs and 20 miRNAs were up-regulated in ovary, 14,142 mRNAs and 12 miRNAs were down-regulated. In DEGs, the known genes related to sex determinism and/or differentiation were also identified, such as DMRT1, SOX9, SF1 for males, FOXL2 for females, and other potentially significant candidate genes. Three sex-related pathways have also been identified, which are Wnt, Notch, and TGF-beta. In 32 DEMs, the three miRNAs (miR-9-5p, miR-92, miR-184) were paid more attention, they predicted 28 target genes, which may also be candidates for sex-related miRNAs and genes. Differential miRNAs target genes analysis reveals the pathway associated with oocyte meiosis and spermatogenesis. Overall, the findings of the study provide significant insights to enhance our understanding of sex differentiation and/or sex determination mechanisms for H. cumingii.
Collapse
Affiliation(s)
- Ya-Yu Wang
- grid.412514.70000 0000 9833 2433Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, 999 Huchenghuan Road, Shanghai, 201306 China ,National Demonstration Center for Experimental Fisheries Science Education, Shanghai, 201306 China ,Shanghai Engineering Research Center of Aquaculture, Shanghai, 201306 China
| | - Sheng-Hua Duan
- grid.412514.70000 0000 9833 2433Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, 999 Huchenghuan Road, Shanghai, 201306 China ,National Demonstration Center for Experimental Fisheries Science Education, Shanghai, 201306 China ,Shanghai Engineering Research Center of Aquaculture, Shanghai, 201306 China
| | - Gui-Ling Wang
- grid.412514.70000 0000 9833 2433Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, 999 Huchenghuan Road, Shanghai, 201306 China ,National Demonstration Center for Experimental Fisheries Science Education, Shanghai, 201306 China ,Shanghai Engineering Research Center of Aquaculture, Shanghai, 201306 China
| | - Jia-Le Li
- grid.412514.70000 0000 9833 2433Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, 999 Huchenghuan Road, Shanghai, 201306 China ,National Demonstration Center for Experimental Fisheries Science Education, Shanghai, 201306 China ,Shanghai Engineering Research Center of Aquaculture, Shanghai, 201306 China
| |
Collapse
|
9
|
Jin Y, Yang M, Gao C, Yue W, Liang X, Xie B, Zhu X, Fan S, Li R, Li M. Fbxo30 regulates chromosome segregation of oocyte meiosis. Cell Mol Life Sci 2019; 76:2217-2229. [PMID: 30980108 PMCID: PMC11105211 DOI: 10.1007/s00018-019-03038-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 01/14/2019] [Accepted: 02/01/2019] [Indexed: 01/18/2023]
Abstract
As the female gamete, meiotic oocytes provide not only half of the genome but also almost all stores for fertilization and early embryonic development. Because de novo mRNA transcription is absent in oocyte meiosis, protein-level regulations, especially the ubiquitin proteasome system, are more crucial. As the largest family of ubiquitin E3 ligases, Skp1-Cullin-F-box complexes recognize their substrates via F-box proteins with substrate-selected specificity. However, the variety of F-box proteins and their unknown substrates hinder our understanding of their functions. In this report, we find that Fbxo30, a new member of F-box proteins, is enriched in mouse oocytes, and its expression level declines substantially after the metaphase of the first meiosis (MI). Notably, depletion of Fbxo30 causes significant chromosome compaction accompanied by chromosome segregation failure and arrest at the MI stage, and this arrest is not caused by over-activation of spindle assembly checkpoint. Using immunoprecipitation and mass spectrometric analysis, we identify stem-loop-binding protein (SLBP) as a novel substrate of Fbxo30. SLBP overexpression caused by Fbxo30 depletion results in a remarkable overload of histone H3 on chromosomes that excessively condenses chromosomes and inhibits chromosome segregation. Our finding uncovers an unidentified pathway-controlling chromosome segregation and cell progress.
Collapse
Affiliation(s)
- Yimei Jin
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Mo Yang
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Chang Gao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
| | - Wei Yue
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaoling Liang
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Bingteng Xie
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Xiaohui Zhu
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Shangrong Fan
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Rong Li
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China.
| | - Mo Li
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China.
| |
Collapse
|
10
|
Wilburn DB, Feldhoff RC. An annual cycle of gene regulation in the red-legged salamander mental gland: from hypertrophy to expression of rapidly evolving pheromones. BMC DEVELOPMENTAL BIOLOGY 2019; 19:10. [PMID: 31029098 PMCID: PMC6487043 DOI: 10.1186/s12861-019-0190-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/10/2019] [Indexed: 02/06/2023]
Abstract
Background Cell differentiation is mediated by synchronized waves of coordinated expression for hundreds to thousands of genes, and must be regulated to produce complex tissues and phenotypes. For many animal species, sexual selection has driven the development of elaborate male ornaments, requiring sex-specific differentiation pathways. One such male ornament is the pheromone-producing mental gland of the red-legged salamander (Plethodon shermani). Mental gland development follows an annual cycle of extreme hypertrophy, production of pheromones for the ~ 2 month mating season, and then complete resorption before repeating the process in the following year. At the peak of the mating season, the transcriptional and translational machinery of the mental gland are almost exclusively redirected to the synthesis of rapidly evolving pheromones. Of these pheromones, Plethodontid Modulating Factor (PMF) has experienced an unusual history: following gene duplication, the protein coding sequence diversified from positive sexual selection while the untranslated regions have been conserved by purifying selection. The molecular underpinnings that bridge the processes of gland hypertrophy, pheromone synthesis, and conservation of the untranslated regions remain to be determined. Results Using Illumina sequencing, we prepared a de novo transcriptome of the mental gland at six stages of development. Differential expression analysis and immunohistochemistry revealed that the mental gland initially adopts a highly proliferative, almost tumor-like phenotype, followed by a rapid increase in pheromone mRNA and protein. One likely player in this transition is Cold Inducible RNA Binding Protein (CIRBP), which selectively and cooperatively binds the highly conserved PMF 3′ UTR. CIRBP, along with other proteins associated with stress response, have seemingly been co-opted to aid in mental gland development by helping to regulate pheromone synthesis. Conclusions The P. shermani mental gland utilizes a complex system of transcriptional and post-transcriptional gene regulation to facilitate its hypertrophication and pheromone synthesis. The data support the evolutionary interplay of coding and noncoding segments in rapid gene evolution, and necessitate the study of co-evolution between pheromone gene products and their transcriptional/translational regulators. Additionally, the mental gland could be a powerful emerging model of regulated tissue proliferation and subsequent resorption within the dermis and share molecular links to skin cancer biology. Electronic supplementary material The online version of this article (10.1186/s12861-019-0190-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Damien B Wilburn
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY, 40292, USA. .,Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA.
| | - Richard C Feldhoff
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY, 40292, USA
| |
Collapse
|
11
|
Zhang N, Li X, Liu X, Cao Y, Chen D, Liu X, Wang Q, Du J, Weng J, Ma W. p21-activated kinase 1 activity is required for histone H3 Ser 10 phosphorylation and chromatin condensation in mouse oocyte meiosis. Reprod Fertil Dev 2018; 29:1287-1296. [PMID: 27166635 DOI: 10.1071/rd16026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/06/2016] [Indexed: 01/04/2023] Open
Abstract
p21-activated kinase 1 (Pak1) is essential for a variety of cellular events, including gene transcription, cytoskeletal organisation, cell proliferation and apoptosis. Pak1 is activated upon autophosphorylation on many amino residues; in particular, phosphorylation on Thr423 maintains maximal Pak1 activation. In the present study we investigated the protein expression, subcellular localisation and function of Pak1 phosphorylated on Thr423 (pPak1Thr423) in mouse oocytes. pPak1Thr423 was detected upon meiotic resumption and localised on the condensing chromatin. Thr423 phosphorylation was markedly suppressed by the Pak1 ATP-competitive inhibitor PF-3758309, but not by the allosteric inhibitors IPA-3 (2.5 μM and 10μM) (1, 1'-dithiobis-2-naphthalenol) and TAT-PAK18 (10 μM), which prevent the binding of Pak1 to its upstream activators GTPase Cdc42/Rac and Pak-interacting exchange factor (PIX), respectively, implying that Pak1 activation may be independent of GTPase and PIX in oocyte meiosis. Inhibition of Pak1 activation concomitantly restrained histone H3 phosphorylation on Ser10 and consequently inhibited chromatin condensation; however, this phenotype was reversed by concomitant administration of the Pak1 activator FTY720. The changes in the pattern of expression of phosphorylated extracellular signal-regulated kinase 1/2 in response to PF-3758309 or FTY720 were the same as seen for pPak1Thr423. These results show that activated Pak1 regulates chromatin condensation by promoting H3 Ser10 phosphorylation in oocytes after the resumption of meiotic progression.
Collapse
Affiliation(s)
- Nana Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiuhong Li
- Biospecimen and Clinical Data Repository, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Xiaoyun Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yan Cao
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Dandan Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiaoyu Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Qian Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Juan Du
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jing Weng
- Experimental Center for Basic Medical Teaching, 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
|
12
|
de Melo TP, de Camargo GMF, de Albuquerque LG, Carvalheiro R. Genome-wide association study provides strong evidence of genes affecting the reproductive performance of Nellore beef cows. PLoS One 2017; 12:e0178551. [PMID: 28562680 PMCID: PMC5451131 DOI: 10.1371/journal.pone.0178551] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/15/2017] [Indexed: 12/31/2022] Open
Abstract
Reproductive traits are economically important for beef cattle production; however, these traits are still a bottleneck in indicine cattle since these animals typically reach puberty at older ages when compared to taurine breeds. In addition, reproductive traits are complex phenotypes, i.e., they are controlled by both the environment and many small-effect genes involved in different pathways. In this study, we conducted genome-wide association study (GWAS) and functional analyses to identify important genes and pathways associated with heifer rebreeding (HR) and with the number of calvings at 53 months of age (NC53) in Nellore cows. A total of 142,878 and 244,311 phenotypes for HR and NC53, respectively, and 2,925 animals genotyped with the Illumina Bovine HD panel (Illumina®, San Diego, CA, USA) were used in GWAS applying the weighted single-step GBLUP (WssGBLUP) method. Several genes associated with reproductive events were detected in the 20 most important 1Mb windows for both traits. Significant pathways for HR and NC53 were associated with lipid metabolism and immune processes, respectively. MHC class II genes, detected on chromosome 23 (window 25-26Mb) for NC53, were significantly associated with pregnancy success of Nellore cows. These genes have been proved previously to be associated with reproductive traits such as mate choice in other breeds and species. Our results suggest that genes associated with the reproductive traits HR and NC53 may be involved in embryo development in mammalian species. Furthermore, some genes associated with mate choice may affect pregnancy success in Nellore cattle.
Collapse
Affiliation(s)
- Thaise Pinto de Melo
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP – Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | | | - Lucia Galvão de Albuquerque
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP – Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
- National Council for Scientific and Technological Development (CNPq), Brasília, DF, Brazil
| | - Roberto Carvalheiro
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP – Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
- National Council for Scientific and Technological Development (CNPq), Brasília, DF, Brazil
- * E-mail:
| |
Collapse
|
13
|
Tuttolomondo A, Simonetta I, Pinto A. MicroRNA and receptor mediated signaling pathways as potential therapeutic targets in heart failure. Expert Opin Ther Targets 2016; 20:1287-1300. [PMID: 27409295 DOI: 10.1080/14728222.2016.1212017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Cardiac remodelling is a complex pathogenetic pathway involving genome expression, molecular, cellular, and interstitial changes that cause changes in size, shape and function of the heart after cardiac injury. Areas covered: We will review recent advances in understanding the role of several receptor-mediated signaling pathways and micro-RNAs, in addition to their potential as candidate target pathways in the pathogenesis of heart failure. The myocyte is the main target cell involved in the remodelling process via ischemia, cell necrosis and apoptosis (by means of various receptor pathways), and other mechanisms mediated by micro-RNAs. We will analyze the role of some receptor mediated signaling pathways such as natriuretic peptides, mediators of glycogen synthase kinase 3 and ERK1/2 pathways, beta-adrenergic receptor subtypes and relaxin receptor signaling mechanisms, TNF/TNF receptor family and TWEAK/Fn14 axis, and some micro-RNAs as candidate target pathways in pathogenesis of heart failure. These mediators of receptor-mediated pathways and micro-RNA are the most addressed targets of emerging therapies in modern heart failure treatment strategies. Expert opinion: Future treatment strategies should address mediators involved in multiple steps within heart failure pathogenetic pathways.
Collapse
Affiliation(s)
- Antonino Tuttolomondo
- a U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S) , University of Palermo , Palermo , Italy
| | - Irene Simonetta
- a U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S) , University of Palermo , Palermo , Italy
| | - Antonio Pinto
- a U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S) , University of Palermo , Palermo , Italy
| |
Collapse
|
14
|
Labrecque R, Fournier E, Sirard MA. Transcriptome analysis of bovine oocytes from distinct follicle sizes: Insights from correlation network analysis. Mol Reprod Dev 2016; 83:558-69. [DOI: 10.1002/mrd.22651] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/19/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Rémi Labrecque
- Faculté des sciences de l'Agriculture et de l'Alimentation; Département des Sciences Animales; Centre de Recherche en Biologie de la Reproduction; Pavillon INAF; Université Laval; Québec QC Canada
| | - Eric Fournier
- Faculté des sciences de l'Agriculture et de l'Alimentation; Département des Sciences Animales; Centre de Recherche en Biologie de la Reproduction; Pavillon INAF; Université Laval; Québec QC Canada
| | - Marc-André Sirard
- Faculté des sciences de l'Agriculture et de l'Alimentation; Département des Sciences Animales; Centre de Recherche en Biologie de la Reproduction; Pavillon INAF; Université Laval; Québec QC Canada
| |
Collapse
|
15
|
Clement TM, Inselman AL, Goulding EH, Willis WD, Eddy EM. Disrupting Cyclin Dependent Kinase 1 in Spermatocytes Causes Late Meiotic Arrest and Infertility in Mice. Biol Reprod 2015; 93:137. [PMID: 26490841 DOI: 10.1095/biolreprod.115.134940] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/15/2015] [Indexed: 01/22/2023] Open
Abstract
While cyclin dependent kinase 1 (CDK1) has a critical role in controlling resumption of meiosis in oocytes, its role has not been investigated directly in spermatocytes. Unique aspects of male meiosis led us to hypothesize that its role is different in male meiosis than in female meiosis. We generated a conditional knockout (cKO) of the Cdk1 gene in mouse spermatocytes to test this hypothesis. We found that CDK1-null spermatocytes undergo synapsis, chiasmata formation, and desynapsis as is seen in oocytes. Additionally, CDK1-null spermatocytes relocalize SYCP3 to centromeric foci, express H3pSer10, and initiate chromosome condensation. However, CDK1-null spermatocytes fail to form condensed bivalent chromosomes in prophase of meiosis I and instead are arrested at prometaphase. Thus, CDK1 has an essential role in male meiosis that is consistent with what is known about the role of CDK1 in female meiosis, where it is required for formation of condensed bivalent metaphase chromosomes and progression to the first meiotic division. We found that cKO spermatocytes formed fully condensed bivalent chromosomes in the presence of okadaic acid, suggesting that cKO chromosomes are competent to condense, although they do not do so in vivo. Additionally, arrested cKO spermatocytes exhibited irregular cell shape, irregular large nuclei, and large distinctive nucleoli. These cells persist in the seminiferous epithelium through the next seminiferous epithelial cycle with a lack of stage XII checkpoint-associated cell death. This indicates that CDK1 is required upstream of a checkpoint-associated cell death as well as meiotic metaphase progression in mouse spermatocytes.
Collapse
Affiliation(s)
- Tracy M Clement
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Amy L Inselman
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Eugenia H Goulding
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - William D Willis
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Edward M Eddy
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
16
|
Rusin SF, Schlosser KA, Adamo ME, Kettenbach AN. Quantitative phosphoproteomics reveals new roles for the protein phosphatase PP6 in mitotic cells. Sci Signal 2015; 8:rs12. [PMID: 26462736 DOI: 10.1126/scisignal.aab3138] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Protein phosphorylation is an important regulatory mechanism controlling mitotic progression. Protein phosphatase 6 (PP6) is an essential enzyme with conserved roles in chromosome segregation and spindle assembly from yeast to humans. We applied a baculovirus-mediated gene silencing approach to deplete HeLa cells of the catalytic subunit of PP6 (PP6c) and analyzed changes in the phosphoproteome and proteome in mitotic cells by quantitative mass spectrometry-based proteomics. We identified 408 phosphopeptides on 272 proteins that increased and 298 phosphopeptides on 220 proteins that decreased in phosphorylation upon PP6c depletion in mitotic cells. Motif analysis of the phosphorylated sites combined with bioinformatics pathway analysis revealed previously unknown PP6c-dependent regulatory pathways. Biochemical assays demonstrated that PP6c opposed casein kinase 2-dependent phosphorylation of the condensin I subunit NCAP-G, and cellular analysis showed that depletion of PP6c resulted in defects in chromosome condensation and segregation in anaphase, consistent with dysregulation of condensin I function in the absence of PP6 activity.
Collapse
Affiliation(s)
- Scott F Rusin
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Kate A Schlosser
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Mark E Adamo
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Arminja N Kettenbach
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA.
| |
Collapse
|
17
|
Iwamoto D, Yamagata K, Kishi M, Hayashi-Takanaka Y, Kimura H, Wakayama T, Saeki K. Early development of cloned bovine embryos produced from oocytes enucleated by fluorescence metaphase II imaging using a conventional halogen-lamp microscope. Cell Reprogram 2015; 17:106-14. [PMID: 25826723 DOI: 10.1089/cell.2014.0086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Enucleation of a recipient oocyte is one of the key processes in the procedure of somatic cell nuclear transfer (SCNT). However, especially in bovine species, lipid droplets spreading in the ooplasm hamper identification and enucleation of metaphase II (MII) chromosomes, and thereby the success rate of the cloning remains low. In this study we used a new experimental system that enables fluorescent observation of chromosomes in living oocytes without any damage. We succeeded in visualizing and removing the MII chromosome in matured bovine oocytes. This experimental system consists of injecting fluorescence-labeled antibody conjugates that bind to chromosomes and fluorescent observation using a conventional halogen-lamp microscope. The cleavage rates and blastocyst rates of bovine embryos following in vitro fertilization (IVF) decreased as the concentration of the antibody increased (p<0.05). The enucleation rate of the conventional method (blind enucleation) was 86%, whereas all oocytes injected with the antibody conjugates were enucleated successfully. Fusion rates and developmental rates of SCNT embryos produced with the enucleated oocytes were the same as those of the blind enucleation group (p>0.05). For the production of SCNT embryos, the new system can be used as a reliable predictor of the location of metaphase plates in opaque oocytes, such as those in ruminant animals.
Collapse
Affiliation(s)
- Daisaku Iwamoto
- 1 Department of Genetic Engineering, Kinki University , Kinokawa, Wakayama, 649-6493, Japan
| | | | | | | | | | | | | |
Collapse
|
18
|
Monteiro da Rocha A, Ding J, Slawny N, Wolf AM, Smith GD. Loss of glycogen synthase kinase 3 isoforms during murine oocyte growth induces offspring cardiac dysfunction. Biol Reprod 2015; 92:127. [PMID: 25833158 DOI: 10.1095/biolreprod.115.128181] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/19/2015] [Indexed: 12/22/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK3) is a constitutively active serine threonine kinase with 1) two isoforms (GSK3A and GSK3B) that have unique and overlapping functions, 2) multiple molecular intracellular mechanisms that involve phosphorylation of diverse substrates, and 3) implications in pathogenesis of many diseases. Insulin causes phosphorylation and inactivation of GSK3 and mammalian oocytes have a functional insulin-signaling pathway whereby prolonged elevated insulin during follicle/oocyte development causes GSK3 hyperphosphorylation, reduced GSK3 activity, and altered oocyte chromatin remodeling. Periconceptional diabetes and chronic hyperinsulinemia are associated with congenital malformations and onset of adult diseases of cardiovascular origin. Objectives were to produce transgenic mice with individual or concomitant loss of GSK3A and/or GSK3B and investigate the in vivo role of oocyte GSK3 on fertility, fetal development, and offspring health. Wild-type males bred to females with individual or concomitant loss of oocyte GSK3 isoforms did not have reduced fertility. However, concomitant loss of GSK3A and GSK3B in the oocyte significantly increased neonatal death rate due to congestive heart failure secondary to ventricular hyperplasia. Individual loss of oocyte GSK3A or GSK3B did not induce this lethal phenotype. In conclusion, absence of oocyte GSK3 in the periconceptional period does not alter fertility yet causes offspring cardiac hyperplasia, cardiovascular defects, and significant neonatal death. These results support a developmental mechanism by which periconceptional hyperinsulinemia associated with maternal metabolic syndrome, obesity, and/or diabetes can act on the oocyte and affect offspring cardiovascular development, function, and congenital heart malformation.
Collapse
Affiliation(s)
| | - Jun Ding
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Nicole Slawny
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Amber M Wolf
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gary D Smith
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan Department of Urology, University of Michigan, Ann Arbor, Michigan Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
19
|
Kang H, Park YS, Cho DH, Kim JS, Oh JS. Dynamics of histone H3 phosphorylation at threonine 3 during meiotic maturation in mouse oocytes. Biochem Biophys Res Commun 2015; 458:280-6. [PMID: 25645018 DOI: 10.1016/j.bbrc.2015.01.099] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 01/21/2015] [Indexed: 11/18/2022]
Abstract
Various histone residues are post-translationally modified during the cell cycle. Among these, histone H3 phosphorylation at threonine 3 (H3T3ph) is newly characterized and has been considered to be crucial for chromosome dynamics during mitosis. However, little is known about the role of H3T3ph during mouse oocyte maturation. In the present study, we examined H3T3ph expression and localization during oocyte meiosis. Our results showed that H3T3ph was tightly associated with condensed chromosomes during meiotic maturation. H3T3ph along the chromosome arms was dissociated at anaphase/telophase I, but centromeric H3T3ph remained intact. Moreover, the inhibition of H3T3ph with the small molecule inhibitors CHR-6494 and 5-Itu impaired segregation of homologous chromosomes during meiosis. Partial inhibition of H3T3ph revealed that centromeric Aurora B/C kinase is sufficient to complete meiosis I, but Aurora B/C kinase along the chromosome arms is required to ensure accurate homologous chromosome segregation. Therefore, our results demonstrate that H3T3ph is a universal regulator of chromosome dynamics during oocyte meiosis and mitosis.
Collapse
Affiliation(s)
- Hyoeun Kang
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Yong Seok Park
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Dong-Hyung Cho
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, South Korea
| | - Jae-Sung Kim
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea.
| | - Jeong Su Oh
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
20
|
Wang B, Ma W, Xu X, Wang C, Zhu Y, An N, An L, Wu Z, Tian J. Phosphorylation of histone H3 on Ser10 by auto-phosphorylated PAK1 is not essential for chromatin condensation and meiotic progression in porcine oocytes. J Anim Sci Biotechnol 2013; 4:13. [PMID: 23521812 PMCID: PMC3639857 DOI: 10.1186/2049-1891-4-13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 03/01/2013] [Indexed: 11/10/2022] Open
Abstract
Background The p21-activated kinase 1 (PAK1) is essential for mitosis and plays an important role in the regulation of microtubule assembly during oocyte meiotic maturation in mice; however, little is known about its role in porcine oocytes. Result Total p21-activated kinase 1 (PAK1) and phosphorylated PAK1 at Thr423 (PAK1Thr423) were consistently expressed in porcine oocytes from the germinal vesicle (GV) to the second metaphase (MII) stages, but phosphorylation of histone H3 at Ser10 (H3Ser10) was only expressed after the GV stage. Immunofluorescence analysis revealed that PAK1Thr423 and H3Ser10 colocalized on chromosomes after the GV stage. Blocking of endogenous PAK1Thr423 by injecting a specific antibody decreased the phosphorylation level of H3Ser10; however, it had no impact on chromatin condensation, meiotic progression, cleavage rate of blastomeres or the rate of blastocyst formation. Conclusion Phosphorylation of PAK1Thr423 is a spontaneous activation process and the activated PAK1Thr423 can promote the phosphorylation of H3Ser10; however, this pathway is not required for meiotic maturation of porcine oocytes or early embryonic development.
Collapse
Affiliation(s)
- Bingyuan Wang
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Wei Ma
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,Department of Histology and Embryology; School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, P. R. China
| | - Xiaoling Xu
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,Institute of Animal Husbandry and Veterinary Medicine, Beijing Municipal Academy of Agriculture and Forestry Sciences, Beijing, 100097, P. R. China
| | - Chao Wang
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Yubo Zhu
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Na An
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Lei An
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Zhonghong Wu
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Jianhui Tian
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| |
Collapse
|
21
|
Ribeiro-Mason K, Boulesteix C, Fleurot R, Aguirre-Lavin T, Adenot P, Gall L, Debey P, Beaujean N. H3S10 phosphorylation marks constitutive heterochromatin during interphase in early mouse embryos until the 4-cell stage. J Reprod Dev 2012; 58:467-75. [PMID: 22572731 DOI: 10.1262/jrd.11-109h] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Phosphorylation of histone H3 at Ser10 (H3S10P) has been linked to a variety of cellular processes, such as chromosome condensation and gene activation/silencing. Remarkably, in mammalian somatic cells, H3S10P initiates in the pericentromeric heterochromatin during the late G2 phase, and phosphorylation spreads throughout the chromosomes arms in prophase, being maintained until the onset of anaphase when it gets dephosphorylated. Considerable studies have been carried out about H3S10P in different organisms; however, there is little information about this histone modification in mammalian embryos. We hypothesized that this epigenetic modification could also be a marker of pericentromeric heterochromatin in preimplantation embryos. We therefore followed the H3S10P distribution pattern in the G1/S and G2 phases through the entire preimplantation development in in vivo mouse embryos. We paid special attention to its localization relative to another pericentromeric heterochromatin marker, HP1β and performed immunoFISH using specific pericentromeric heterochromatin probes. Our results indicate that H3S10P presents a remarkable distribution pattern in preimplantation mouse embryos until the 4-cell stage and is a better marker of pericentromeric heterochromatin than HP1β. After the 8-cell stage, H3S10P kinetic is more similar to the somatic one, initiating during G2 in chromocenters and disappearing upon telophase. Based on these findings, we believe that H3S10P is a good marker of pericentromeric heterochromatin, especially in the late 1- and 2-cell stages as it labels both parental genomes and that it can be used to further investigate epigenetic regulation and heterochromatin mechanisms in early preimplantation embryos.
Collapse
Affiliation(s)
- Karlla Ribeiro-Mason
- INRA, UMR 1198 Biologie du Développement et Reproduction, F-78350 Jouy en Josas, France
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Current advances in epigenetic modification and alteration during mammalian ovarian folliculogenesis. J Genet Genomics 2012; 39:111-23. [PMID: 22464470 DOI: 10.1016/j.jgg.2012.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 01/07/2012] [Accepted: 02/10/2012] [Indexed: 11/23/2022]
Abstract
During the growth and development of mammalian ovarian follicles, the activation and deactivation of mass genes are under the synergistic control of diverse modifiers through genetic and epigenetic events. Many factors regulate gene activity and functions through epigenetic modification without altering the DNA sequence, and the common mechanisms may include but are not limited to: DNA methylation, histone modifications (e.g., acetylation, deacetylation, phosphorylation, methylation, and ubiquitination), and RNA-associated silencing of gene expression by noncoding RNA. Over the past decade, substantial progress has been achieved in studies involving the epigenetic alterations during mammalian germ cell development. A number of candidate regulatory factors have been identified. This review focuses on the current available information of epigenetic alterations (e.g., DNA methylation, histone modification, noncoding-RNA-mediated regulation) during mammalian folliculogenesis and recounts when and how epigenetic patterns are differentially established, maintained, or altered in this process. Based on different types of epigenetic regulation, our review follows the temporal progression of events during ovarian folliculogenesis and describes the epigenetic changes and their contributions to germ cell-specific functions at each stage (i.e., primordial folliculogenesis (follicle formation), follicle maturation, and follicular atresia).
Collapse
|
23
|
Wang ZB, Schatten H, Sun QY. Why is chromosome segregation error in oocytes increased with maternal aging? Physiology (Bethesda) 2012; 26:314-25. [PMID: 22013190 DOI: 10.1152/physiol.00020.2011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
It is well documented that female fertility is decreased with advanced maternal age due to chromosome abnormality in oocytes. Increased chromosome missegregation is mainly caused by centromeric cohesion reduction. Other factors such as weakened homologous recombination, improper spindle organization, spindle assembly checkpoint (SAC) malfunction, chromatin epigenetic changes, and extra-oocyte factors may also cause chromosome errors.
Collapse
Affiliation(s)
- Zhen-Bo Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | |
Collapse
|
24
|
Chang HY, Jennings PC, Stewart J, Verrills NM, Jones KT. Essential role of protein phosphatase 2A in metaphase II arrest and activation of mouse eggs shown by okadaic acid, dominant negative protein phosphatase 2A, and FTY720. J Biol Chem 2011; 286:14705-12. [PMID: 21383018 DOI: 10.1074/jbc.m110.193227] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vertebrate eggs arrest at second meiotic metaphase. The fertilizing sperm causes meiotic exit through Ca(2+)-mediated activation of the anaphase-promoting complex/cyclosome (APC/C). Although the loss in activity of the M-phase kinase CDK1 is known to be an essential downstream event of this process, the contribution of phosphatases to arrest and meiotic resumption is less apparent, especially in mammals. Therefore, we explored the role of protein phosphatase 2A (PP2A) in mouse eggs using pharmacological inhibition and activation as well as a functionally dominant-negative catalytic PP2A subunit (dn-PP2Ac-L199P) coupled with live cell imaging. We observed that PP2A inhibition using okadaic acid induced events normally observed at fertilization: degradation of the APC/C substrates cyclin B1 and securin resulting from loss of the APC/C inhibitor Emi2. Although sister chromatids separated, chromatin remained condensed, and polar body extrusion was blocked as a result of a rapid spindle disruption, which could be ameliorated by non-degradable cyclin B1, suggesting that spindle integrity was affected by CDK1 loss. Similar cell cycle effects to okadaic acid were also observed using dominant-negative PP2Ac. Preincubation of eggs with the PP2A activator FTY720 could block many of the actions of okadaic acid, including Emi2, cyclin B1, and securin degradation and sister chromatid separation. Therefore, in conclusion, we used okadaic acid, dn-PP2Ac-L199P, and FTY720 on mouse eggs to demonstrate that PP2A is needed to for both continued metaphase arrest and successful exit from meiosis.
Collapse
Affiliation(s)
- Heng-Yu Chang
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, 2308, Australia
| | | | | | | | | |
Collapse
|
25
|
Ding J, Swain JE, Smith GD. Aurora kinase-A regulates microtubule organizing center (MTOC) localization, chromosome dynamics, and histone-H3 phosphorylation in mouse oocytes. Mol Reprod Dev 2011; 78:80-90. [PMID: 21274965 DOI: 10.1002/mrd.21272] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 12/14/2010] [Indexed: 01/08/2023]
Abstract
Aurora kinases (AURKs) are conserved serine/threonine kinases, crucial in regulating cell cycle events. Mammalian oocytes express all three Aurk isoforms throughout meiosis, with AurkA being the predominant isoform. Inhibition of all AURK isoforms by pharmacological means disrupts oocyte meiosis. Therefore, AurkA short interfering RNA (siRNA) was performed to silence AurkA gene expression in mouse oocytes and to further assess the function of AurkA during meiosis by analyzing subsequent loss-of-function oocyte phenotypes. Results indicated that AurkA siRNA applied in our experiments specifically knocked down both AurkA gene and protein expression without influencing transcript levels of AurkB/AurkC and other endogenous protein expression, such as GAPDH and ERK-2. AURKA was not essential for resumption of meiosis, but it potentiated oocyte meiotic progression. Knockdown of AurkA led to a significant reduction in the number of oocytes proceeding to metaphase II (MII). AurkA siRNA resulted in abnormal spindle assembly, improper localization of microtubule organizing centers (MTOCs) and misalignment of chromosomes in metaphase I (MI) oocytes. Co-immunoprecipitations demonstrated that AURKA was physically associated with phospho-Histone H3 ser10 in meiotic oocytes. AurkA siRNA dramatically reduced Histone H3 ser10 phosphorylation, but not ser28, and resulted in a significant increase of abnormal chromosome segregation in MII oocytes. In conclusion, as a predominant isoform among Aurks in oocytes, AurkA plays critical roles in mouse oocyte meiosis by regulating spindle and chromosome dynamics.
Collapse
Affiliation(s)
- Jun Ding
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
26
|
Schober CS, Aydiner F, Booth CJ, Seli E, Reinke V. The kinase VRK1 is required for normal meiotic progression in mammalian oogenesis. Mech Dev 2011; 128:178-90. [PMID: 21277975 DOI: 10.1016/j.mod.2011.01.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 01/19/2011] [Accepted: 01/20/2011] [Indexed: 01/02/2023]
Abstract
The kinase VRK1 has been implicated in mitotic and meiotic progression in invertebrate species, but whether it mediates these events during mammalian gametogenesis is not completely understood. Previous work has demonstrated a role for mammalian VRK1 in proliferation of male spermatogonia, yet whether VRK1 plays a role in meiotic progression, as seen in Drosophila, has not been determined. Here, we have established a mouse strain bearing a gene trap insertion in the VRK1 locus that disrupts Vrk1 expression. In addition to the male proliferation defects, we find that reduction of VRK1 activity causes a delay in meiotic progression during oogenesis, results in the presence of lagging chromosomes during formation of the metaphase plate, and ultimately leads to the failure of oocytes to be fertilized. The activity of at least one phosphorylation substrate of VRK1, p53, is not required for these defects. These results are consistent with previously defined functions of VRK1 in meiotic progression in Drosophila oogenesis, and indicate a conserved role for VRK1 in coordinating proper chromosomal configuration in female meiosis.
Collapse
Affiliation(s)
- Carolyn S Schober
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | |
Collapse
|
27
|
Kan R, Yurttas P, Kim B, Jin M, Wo L, Lee B, Gosden R, Coonrod SA. Regulation of mouse oocyte microtubule and organelle dynamics by PADI6 and the cytoplasmic lattices. Dev Biol 2010; 350:311-22. [PMID: 21147087 DOI: 10.1016/j.ydbio.2010.11.033] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 10/22/2010] [Accepted: 11/29/2010] [Indexed: 11/18/2022]
Abstract
Organelle positioning and movement in oocytes is largely mediated by microtubules (MTs) and their associated motor proteins. While yet to be studied in germ cells, cargo trafficking in somatic cells is also facilitated by specific recognition of acetylated MTs by motor proteins. We have previously shown that oocyte-restricted PADI6 is essential for formation of a novel oocyte-restricted fibrous structure, the cytoplasmic lattices (CPLs). Here, we show that α-tubulin appears to be associated with the PADI6/CPL complex. Next, we demonstrate that organelle positioning and redistribution is defective in PADI6-null oocytes and that alteration of MT polymerization or MT motor activity does not induce organelle redistribution in these oocytes. Finally, we report that levels of acetylated microtubules are dramatically suppressed in the cytoplasm of PADI6-null oocytes, suggesting that the observed organelle redistribution failure is due to defects in stable cytoplasmic MTs. These results demonstrate that the PADI6/CPL superstructure plays a key role in regulating MT-mediated organelle positioning and movement.
Collapse
Affiliation(s)
- Rui Kan
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Baumann C, Viveiros MM, De La Fuente R. Loss of maternal ATRX results in centromere instability and aneuploidy in the mammalian oocyte and pre-implantation embryo. PLoS Genet 2010; 6:e1001137. [PMID: 20885787 PMCID: PMC2944790 DOI: 10.1371/journal.pgen.1001137] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 08/24/2010] [Indexed: 01/10/2023] Open
Abstract
The α-thalassemia/mental retardation X-linked protein (ATRX) is a chromatin-remodeling factor known to regulate DNA methylation at repetitive sequences of the human genome. We have previously demonstrated that ATRX binds to pericentric heterochromatin domains in mouse oocytes at the metaphase II stage where it is involved in mediating chromosome alignment at the meiotic spindle. However, the role of ATRX in the functional differentiation of chromatin structure during meiosis is not known. To test ATRX function in the germ line, we developed an oocyte-specific transgenic RNAi knockdown mouse model. Our results demonstrate that ATRX is required for heterochromatin formation and maintenance of chromosome stability during meiosis. During prophase I arrest, ATRX is necessary to recruit the transcriptional regulator DAXX (death domain associated protein) to pericentric heterochromatin. At the metaphase II stage, transgenic ATRX-RNAi oocytes exhibit abnormal chromosome morphology associated with reduced phosphorylation of histone 3 at serine 10 as well as chromosome segregation defects leading to aneuploidy and severely reduced fertility. Notably, a large proportion of ATRX-depleted oocytes and 1-cell stage embryos exhibit chromosome fragments and centromeric DNA–containing micronuclei. Our results provide novel evidence indicating that ATRX is required for centromere stability and the epigenetic control of heterochromatin function during meiosis and the transition to the first mitosis. The transmission of an abnormal chromosome complement from the gametes to the early embryo, a condition called aneuploidy, is a major cause of congenital birth defects and pregnancy loss. Human embryos are particularly susceptible to aneuploidy, which in the majority of cases is the result of abnormal meiosis in the female gamete. However, the molecular mechanisms involved in the onset of aneuploidy in mammalian oocytes are not fully understood. We show here that, the α-thalassemia/mental retardation X-linked protein (ATRX) is essential for the maintenance of chromosome stability during female meiosis. ATRX is required to recruit the transcriptional regulator DAXX to pericentric heterochromatin at prophase I of meiosis. Notably, lack of ATRX function at the metaphase II stage interferes with the establishment of chromatin modifications associated with chromosome condensation leading to segregation defects, chromosome fragmentation, and severely reduced fertility. Our results provide direct evidence for a role of ATRX in the regulation of pericentric heterochromatin structure and function in mammalian oocytes and have important implications for our understanding of the epigenetic factors contributing to the onset of aneuploidy in the female gamete.
Collapse
Affiliation(s)
- Claudia Baumann
- Female Germ Cell Biology Group, Department of Clinical Studies, University of Pennsylvania, Kennett Square, Pennsylvania, United States of America
| | - Maria M. Viveiros
- Department of Animal Biology, Center for Animal Transgenesis and Germ Cell Research, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania, United States of America
| | - Rabindranath De La Fuente
- Female Germ Cell Biology Group, Department of Clinical Studies, University of Pennsylvania, Kennett Square, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
29
|
Impact of marine drugs on cytoskeleton-mediated reproductive events. Mar Drugs 2010; 8:881-915. [PMID: 20479959 PMCID: PMC2866467 DOI: 10.3390/md8040881] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 03/02/2010] [Accepted: 03/23/2010] [Indexed: 12/30/2022] Open
Abstract
Marine organisms represent an important source of novel bioactive compounds, often showing unique modes of action. Such drugs may be useful tools to study complex processes such as reproduction; which is characterized by many crucial steps that start at gamete maturation and activation and virtually end at the first developmental stages. During these processes cytoskeletal elements such as microfilaments and microtubules play a key-role. In this review we describe: (i) the involvement of such structures in both cellular and in vitro processes; (ii) the toxins that target the cytoskeletal elements and dynamics; (iii) the main steps of reproduction and the marine drugs that interfere with these cytoskeleton-mediated processes. We show that marine drugs, acting on microfilaments and microtubules, exert a wide range of impacts on reproductive events including sperm maturation and motility, oocyte maturation, fertilization, and early embryo development.
Collapse
|
30
|
Rao N, Jhamb D, Milner DJ, Li B, Song F, Wang M, Voss SR, Palakal M, King MW, Saranjami B, Nye HLD, Cameron JA, Stocum DL. Proteomic analysis of blastema formation in regenerating axolotl limbs. BMC Biol 2009; 7:83. [PMID: 19948009 PMCID: PMC2794268 DOI: 10.1186/1741-7007-7-83] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 11/30/2009] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Following amputation, urodele salamander limbs reprogram somatic cells to form a blastema that self-organizes into the missing limb parts to restore the structure and function of the limb. To help understand the molecular basis of blastema formation, we used quantitative label-free liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS)-based methods to analyze changes in the proteome that occurred 1, 4 and 7 days post amputation (dpa) through the mid-tibia/fibula of axolotl hind limbs. RESULTS We identified 309 unique proteins with significant fold change relative to controls (0 dpa), representing 10 biological process categories: (1) signaling, (2) Ca2+ binding and translocation, (3) transcription, (4) translation, (5) cytoskeleton, (6) extracellular matrix (ECM), (7) metabolism, (8) cell protection, (9) degradation, and (10) cell cycle. In all, 43 proteins exhibited exceptionally high fold changes. Of these, the ecotropic viral integrative factor 5 (EVI5), a cell cycle-related oncoprotein that prevents cells from entering the mitotic phase of the cell cycle prematurely, was of special interest because its fold change was exceptionally high throughout blastema formation. CONCLUSION Our data were consistent with previous studies indicating the importance of inositol triphosphate and Ca2+ signaling in initiating the ECM and cytoskeletal remodeling characteristic of histolysis and cell dedifferentiation. In addition, the data suggested that blastema formation requires several mechanisms to avoid apoptosis, including reduced metabolism, differential regulation of proapoptotic and antiapoptotic proteins, and initiation of an unfolded protein response (UPR). Since there is virtually no mitosis during blastema formation, we propose that high levels of EVI5 function to arrest dedifferentiated cells somewhere in the G1/S/G2 phases of the cell cycle until they have accumulated under the wound epidermis and enter mitosis in response to neural and epidermal factors. Our findings indicate the general value of quantitative proteomic analysis in understanding the regeneration of complex structures.
Collapse
Affiliation(s)
- Nandini Rao
- Department of Biology and Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Deepali Jhamb
- School of Informatics and Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Derek J Milner
- Department of Cell and Developmental Biology, and Regeneration Biology and Tissue Engineering Theme, Institute for Genomic Biology, University of Illinois-Urbana Champaign, Urbana, IL, USA
| | - Bingbing Li
- Department of Biology and Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Fengyu Song
- Department of Oral Biology, School of Dentistry and Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Mu Wang
- Department of Biochemistry, School of Medicine and Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - S Randal Voss
- Department of Biology and Spinal Cord and Brain Injury Center, University of Kentucky at Lexington, Lexington, KY, USA
| | - Mathew Palakal
- School of Informatics and Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Michael W King
- Department of Biochemistry, School of Medicine and Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Behnaz Saranjami
- Department of Biology and Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Holly LD Nye
- Department of Cell and Developmental Biology, and Regeneration Biology and Tissue Engineering Theme, Institute for Genomic Biology, University of Illinois-Urbana Champaign, Urbana, IL, USA
| | - Jo Ann Cameron
- Department of Cell and Developmental Biology, and Regeneration Biology and Tissue Engineering Theme, Institute for Genomic Biology, University of Illinois-Urbana Champaign, Urbana, IL, USA
| | - David L Stocum
- Department of Biology and Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
31
|
Silvestre F, Tosti E. Impact of marine drugs on animal reproductive processes. Mar Drugs 2009; 7:539-64. [PMID: 20098597 PMCID: PMC2810222 DOI: 10.3390/md7040539] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 11/03/2009] [Accepted: 11/06/2009] [Indexed: 01/09/2023] Open
Abstract
The discovery and description of bioactive substances from natural sources has been a research topic for the last 50 years. In this respect, marine animals have been used to extract many new compounds exerting different actions. Reproduction is a complex process whose main steps are the production and maturation of gametes, their activation, the fertilisation and the beginning of development. In the literature it has been shown that many substances extracted from marine organisms may have profound influence on the reproductive behaviour, function and reproductive strategies and survival of species. However, despite the central importance of reproduction and thus the maintenance of species, there are still few studies on how reproductive mechanisms are impacted by marine bioactive drugs. At present, studies in either marine and terrestrial animals have been particularly important in identifying what specific fine reproductive mechanisms are affected by marine-derived substances. In this review we describe the main steps of the biology of reproduction and the impact of substances from marine environment and organisms on the reproductive processes.
Collapse
Affiliation(s)
| | - Elisabetta Tosti
- Author to whom correspondence should be addressed; E-Mail:
; Tel.: +39 081 5833288; Fax: +39 081 7641355
| |
Collapse
|
32
|
Sun F, Handel MA. Regulation of the meiotic prophase I to metaphase I transition in mouse spermatocytes. Chromosoma 2008; 117:471-85. [PMID: 18563426 PMCID: PMC2737826 DOI: 10.1007/s00412-008-0167-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Revised: 04/21/2008] [Accepted: 05/05/2008] [Indexed: 11/26/2022]
Abstract
The meiotic prophase I to metaphase I transition (G2/MI) involves disassembly of synaptonemal complex (SC), chromatin condensation, and final compaction of morphologically distinct MI bivalent chromosomes. Control of these processes is poorly understood. The G2/MI transition was experimentally induced in mouse pachytene spermatocytes by okadaic acid (OA), and kinetic analysis revealed that disassembly of the central element of the SC occurred very rapidly after OA treatment, before histone H3 phosphorylation on Ser10. These events were followed by relocalization of SYCP3 and final condensation of bivalents. Enzymatic control of these G2/MI transition events was studied using small molecule inhibitors: butyrolactone I (BLI), an inhibitor of cyclin-dependent kinases (CDKs) and ZM447439 (ZM), an inhibitor of aurora kinases (AURKs). The formation of highly condensed MI bivalents and disassembly of the SC are regulated by both CDKs and AURKs. AURKs also mediate phosphorylation of histone H3 in meiosis. However, neither BLI nor ZM inhibited disassembly of the central element of the SC. Thus, despite evidence that the metaphase promoting factor is a universal regulator of the onset of cell division, desynapsis, the first and key step of the G2/MI transition, occurs independently of BLI-sensitive CDKs and ZM-sensitive AURKs.
Collapse
Affiliation(s)
- Fengyun Sun
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | |
Collapse
|
33
|
Kramer JM, Evans A, Drury K, Moore K. Calyculin-A improves chromosome condensation for cytogenetic analysis of blastomeres from bovine and murine eight-cell stage embryos. Theriogenology 2008; 70:692-703. [PMID: 18556054 DOI: 10.1016/j.theriogenology.2008.04.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2007] [Revised: 04/21/2008] [Accepted: 04/23/2008] [Indexed: 11/25/2022]
Abstract
This study evaluated the serine/threonine phosphatase inhibitor calyculin-A for rapid, efficient induction of premature chromosome condensation (PCC) in blastomeres obtained from Day 3 bovine and Day 2 murine eight-cell stage embryos, and its potential for use in cytogenetic analysis. Experiment 1 tested calyculin-A duration (0, 60, 120, and 180min) to induce PCC in bovine blastomeres. More blastomeres that underwent PCC had chromosomes suitable for cytogenetic analysis if treated for 120 or 180min (P<0.005). Experiment 2 compared doses of calyculin-A (0, 10, 50, and 100nM) on bovine blastomeres; calyculin-A (50nM, 120min) induced PCC suitable for cytogenetic analysis in the greatest number of blastomeres when compared to other doses (52.5%; P<0.005). Effects of calyculin-A (50nM) on murine blastomeres at durations of 0, 60, 90, and 120min to induce PCC were tested in Experiment 3, with 90min inducing the highest frequency of condensed chromosomes suitable for cytogenetic analysis (34%; P<0.05). Finally, Experiment 4 evaluated calyculin-A treated bovine embryos under optimal conditions (50nM, 120min) for use in gender and cytogenetic analysis. Whole chromosome paint probes were successfully hybridized to chromosomes along with 4',6-diamidino-2-phenylindole dihydrochloride hydrate (DAPI) counterstaining, allowing detection of embryo gender (54% F:46% M) and ploidy of individual blastomeres within embryos (64% diploid:36% mixoploid embryos). In conclusion, we inferred that calyculin-A was useful for rapid induction of PCC, producing chromosome spreads suitable for cytogenetic analysis of blastomeres in G1 or G2/M phase of the cell cycle.
Collapse
Affiliation(s)
- J M Kramer
- Department of Animal Sciences, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| | | | | | | |
Collapse
|
34
|
Swain JE, Ding J, Wu J, Smith GD. Regulation of spindle and chromatin dynamics during early and late stages of oocyte maturation by aurora kinases. Mol Hum Reprod 2008; 14:291-9. [PMID: 18353803 PMCID: PMC2408935 DOI: 10.1093/molehr/gan015] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Examination of factors regulating oocyte chromatin remodeling is crucial to circumvent embryonic aneuploidy and resulting defects. Aurora kinases (AURK) are involved in regulation of chromatin remodeling, however, little attention has been paid to AURKs in regard to oocyte maturation. Meiotically incompetent mouse oocytes contain transcripts for all three Aurk isoforms: A, B and C. Upon achieving meiotic competence, oocytes showed significant increases in transcript levels of all three Aurk isoforms and transcript levels remained unchanged as oocytes progressed through meiosis, with AurkA being the predominant isoform. Inhibition of oocyte AURKs during the prophase–metaphase I (MI) transition via inhibitor ZM447439 (ZM) had no effect on germinal vesicle breakdown. However, meiotic spindles were malformed, and microtubule organizing centers and chromatin were scattered. Chromosomal spreads of MI oocytes indicated AURK inhibition resulted in abnormal chromosome condensation. Furthermore, inhibition of AURK during prophase I–MII prevented completion of MII and extrusion of the polar body. Inhibition of AURKs during the MI–MII transition resulted in significantly fewer cells progressing to MII and induced aberrant chromatin remodeling. Further analysis indicated that inhibition of AURKs resulted in absence of histone-H3 phosphorylation at serine 10 and 28. These data suggest a ZM-sensitive AURK may be an oocyte histone-H3 kinase capable of regulating chromatin remodeling throughout oocyte meiosis, both pre- and post-MI.
Collapse
Affiliation(s)
- Jason E Swain
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
35
|
Ducibella T, Fissore R. The roles of Ca2+, downstream protein kinases, and oscillatory signaling in regulating fertilization and the activation of development. Dev Biol 2008; 315:257-79. [PMID: 18255053 DOI: 10.1016/j.ydbio.2007.12.012] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Revised: 12/12/2007] [Accepted: 12/13/2007] [Indexed: 12/12/2022]
Abstract
Reviews in Developmental Biology have covered the pathways that generate the all-important intracellular calcium (Ca(2+)) signal at fertilization [Miyazaki, S., Shirakawa, H., Nakada, K., Honda, Y., 1993a. Essential role of the inositol 1,4,5-trisphosphate receptor/Ca(2+) release channel in Ca(2+) waves and Ca(2+) oscillations at fertilization of mammalian eggs. Dev. Biol. 158, 62-78; Runft, L., Jaffe, L., Mehlmann, L., 2002. Egg activation at fertilization: where it all begins. Dev. Biol. 245, 237-254] and the different temporal responses of Ca(2+) in many organisms [Stricker, S., 1999. Comparative biology of calcium signaling during fertilization and egg activation in animals. Dev. Biol. 211, 157-176]. Those reviews raise the importance of identifying how Ca(2+) causes the events of egg activation (EEA) and to what extent these temporal Ca(2+) responses encode developmental information. This review covers recent studies that have analyzed how these Ca(2+) signals are interpreted by specific proteins, and how these proteins regulate various EEA responsible for the onset of development. Many of these proteins are protein kinases (CaMKII, PKC, MPF, MAPK, MLCK) whose activity is directly or indirectly regulated by Ca(2+), and whose amount increases during late oocyte maturation. We cover biochemical progress in defining the signaling pathways between Ca(2+) and the EEA, as well as discuss how oscillatory or multiple Ca(2+) signals are likely to have specific advantages biochemically and/or developmentally. These emerging concepts are put into historical context, emphasizing that key contributions have come from many organisms. The intricate interdependence of Ca(2+), Ca(2+)-dependent proteins, and the EEA raise many new questions for future investigations that will provide insight into the extent to which fertilization-associated signaling has long-range implications for development. In addition, answers to these questions should be beneficial to establishing parameters of egg quality for human and animal IVF, as well as improving egg activation protocols for somatic cell nuclear transfer to generate stem cells and save endangered species.
Collapse
Affiliation(s)
- Tom Ducibella
- Department of OB/GYN, Tufts-New England Medical Center, Boston, MA 02111, USA.
| | | |
Collapse
|
36
|
Abu-Farha M, Lambert JP, Al-Madhoun AS, Elisma F, Skerjanc IS, Figeys D. The tale of two domains: proteomics and genomics analysis of SMYD2, a new histone methyltransferase. Mol Cell Proteomics 2007; 7:560-72. [PMID: 18065756 DOI: 10.1074/mcp.m700271-mcp200] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Very little is known about SET- and MYND-containing protein 2 (SMYD2), a member of the SMYD protein family. However, the interest in better understanding the roles of SMYD2 has grown because of recent reports indicating that SMYD2 methylates p53 and histone H3. In this study, we present a combined proteomics and genomics study of SMYD2 designed to elucidate its molecular roles. We report the cytosolic and nuclear interactome of SMYD2 using a combination of immunoprecipitation coupled with high throughput MS, chromatin immunoprecipitation coupled with high throughput MS, and co-immunoprecipitation methods. In particular, we report that SMYD2 interacted with HSP90alpha independently of the SET and MYND domains, with EBP41L3 through the MYND domain, and with p53 through the SET domain. We demonstrated that the interaction of SMYD2 with HSP90alpha enhances SMYD2 histone methyltransferase activity and specificity for histone H3 at lysine 4 (H3K4) in vitro. Interestingly histone H3K36 methyltransferase activity was independent of its interaction with HSP90alpha similar to LSD1 dependence on the androgen receptor. We also showed that the SET domain is required for the methylation at H3K4. We demonstrated using a modified chromatin immunoprecipitation protocol that the SMYD2 gain of function leads to an increase in H3K4 methylation in vivo, whereas no observable levels of H3K36 were detected. We also report that the SMYD2 gain of function was correlated with the up-regulation of 37 and down-regulation of four genes, the majority of which are involved in the cell cycle, chromatin remodeling, and transcriptional regulation. TACC2 is one of the genes up-regulated as a result of SMYD2 gain of function. Up-regulation of TACC2 by SMYD2 occurred as a result of SMYD2 binding to the TACC2 promoter where it methylates H3K4. Furthermore the combination of the SMYD2 interactome with the gene expression data suggests that some of the genes regulated by SMYD2 are closely associated with SMYD2-interacting proteins.
Collapse
Affiliation(s)
- Mohamed Abu-Farha
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | | | | | | | | | | |
Collapse
|