1
|
Kretschmer M, Fischer V, Gapp K. When Dad's Stress Gets under Kid's Skin-Impacts of Stress on Germline Cargo and Embryonic Development. Biomolecules 2023; 13:1750. [PMID: 38136621 PMCID: PMC10742275 DOI: 10.3390/biom13121750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Multiple lines of evidence suggest that paternal psychological stress contributes to an increased prevalence of neuropsychiatric and metabolic diseases in the progeny. While altered paternal care certainly plays a role in such transmitted disease risk, molecular factors in the germline might additionally be at play in humans. This is supported by findings on changes to the molecular make up of germ cells and suggests an epigenetic component in transmission. Several rodent studies demonstrate the correlation between paternal stress induced changes in epigenetic modifications and offspring phenotypic alterations, yet some intriguing cases also start to show mechanistic links in between sperm and the early embryo. In this review, we summarise efforts to understand the mechanism of intergenerational transmission from sperm to the early embryo. In particular, we highlight how stress alters epigenetic modifications in sperm and discuss the potential for these modifications to propagate modified molecular trajectories in the early embryo to give rise to aberrant phenotypes in adult offspring.
Collapse
Affiliation(s)
- Miriam Kretschmer
- Laboratory of Epigenetics and Neuroendocrinology, Department of Health Sciences and Technology, Institute for Neuroscience, ETH Zürich, 8057 Zürich, Switzerland; (M.K.); (V.F.)
- Neuroscience Center Zurich, ETH Zürich and University of Zürich, 8057 Zürich, Switzerland
| | - Vincent Fischer
- Laboratory of Epigenetics and Neuroendocrinology, Department of Health Sciences and Technology, Institute for Neuroscience, ETH Zürich, 8057 Zürich, Switzerland; (M.K.); (V.F.)
- Neuroscience Center Zurich, ETH Zürich and University of Zürich, 8057 Zürich, Switzerland
| | - Katharina Gapp
- Laboratory of Epigenetics and Neuroendocrinology, Department of Health Sciences and Technology, Institute for Neuroscience, ETH Zürich, 8057 Zürich, Switzerland; (M.K.); (V.F.)
- Neuroscience Center Zurich, ETH Zürich and University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
2
|
Cui L, Fang L, Zhuang L, Shi B, Lin CP, Ye Y. Sperm-borne microRNA-34c regulates maternal mRNA degradation and preimplantation embryonic development in mice. Reprod Biol Endocrinol 2023; 21:40. [PMID: 37101140 PMCID: PMC10131327 DOI: 10.1186/s12958-023-01089-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 04/05/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Studies have shown that sperm-borne microRNAs (miRNAs) are involved in mammalian preimplantation embryonic development. In humans, spermatozoan miR-34c levels are correlated with in vitro fertilization outcomes, such as embryo quality and the clinical pregnancy and live birth rates. In rabbits and cows, miR-34c improves the developmental competence of embryos generated by somatic cell nuclear transfer. However, the mechanisms underlying the regulation of embryonic development by miR-34c remain unknown. METHODS Female C57BL/6 mice (6-8 weeks old) were superovulated, and pronucleated zygotes were collected and microinjected with an miR-34c inhibitor or a negative-control RNA. The embryonic development of the microinjected zygotes was evaluated, and the messenger RNA (mRNA) expression profiles of the embryos at the two-cell, four-cell and blastocyst stages (five embryos per group) were determined by RNA sequencing analysis. Gene expression levels were verified by reverse transcription-quantitative polymerase chain reaction. Cluster analysis and heat map visualization were performed to detect differentially expressed mRNAs. Pathway and process enrichment analyses were performed using ontology resources. Differentially expressed mRNAs were systematically analyzed using the Search Tool for the Retrieval of Interacting Genes/Proteins database to determine their biological functions. RESULTS Embryonic developmental potential was significantly reduced in zygotes microinjected with the miR-34c inhibitor compared with those microinjected with a negative-control RNA. Two-cell stage embryos microinjected with an miR-34c inhibitor presented altered transcriptomic profiles, with upregulated expression of maternal miR-34c target mRNAs and classical maternal mRNAs. Differentially expressed transcripts were mainly of genes associated with lipid metabolism and cellular membrane function at the two-cell stage, with cell-cycle phase transition and energy metabolism at the four-cell stage; and with vesicle organization, lipid biosynthetic process and endomembrane system organization at the blastocyst stage. We also showed that genes related to preimplantation embryonic development, including Alkbh4, Sp1, Mapk14, Sin3a, Sdc1 and Laptm4b, were significantly downregulated after microinjection of an miR-34c inhibitor. CONCLUSIONS Sperm-borne miR-34c may regulate preimplantation embryonic development by affecting multiple biological processes, such as maternal mRNA degradation, cellular metabolism, cell proliferation and blastocyst implantation. Our data demonstrate the importance of sperm-derived miRNAs in the development of preimplantation embryos.
Collapse
Affiliation(s)
- Long Cui
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Li Fang
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lili Zhuang
- Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200041, China
| | - Biwei Shi
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yinghui Ye
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
3
|
Parental competition for the regulators of chromatin dynamics in mouse zygotes. Commun Biol 2022; 5:699. [PMID: 35835981 PMCID: PMC9283401 DOI: 10.1038/s42003-022-03623-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 06/22/2022] [Indexed: 11/08/2022] Open
Abstract
The underlying mechanism for parental asymmetric chromatin dynamics is still unclear. To reveal this, we investigate chromatin dynamics in parthenogenetic, androgenic, and several types of male germ cells-fertilized zygotes. Here we illustrate that parental conflicting role mediates the regulation of chromatin dynamics. Sperm reduces chromatin dynamics in both parental pronuclei (PNs). During spermiogenesis, male germ cells acquire this reducing ability and its resistance. On the other hand, oocytes can increase chromatin dynamics. Notably, the oocytes-derived chromatin dynamics enhancing ability is dominant for the sperm-derived opposing one. This maternal enhancing ability is competed between parental pronuclei. Delayed fertilization timing is critical for this competition and compromises parental asymmetric chromatin dynamics and zygotic transcription. Together, parental competition for the maternal factor enhancing chromatin dynamics is a determinant to establish parental asymmetry, and paternal repressive effects have supporting roles to enhance asymmetry.
Collapse
|
4
|
Nuclear-to-cytoplasmic ratios of 1PN and 2PN zygotes after in vitro fertilization of mouse oocytes. ZYGOTE 2021; 30:120-124. [PMID: 34180386 DOI: 10.1017/s096719942100040x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Numerous studies have reported comparisons of the nuclear-to-cytoplasmic (NC) ratio during mitosis. However, little information is known about how the pronuclear size is regulated and determined at the end of meiosis II in mammalian zygotes. The present study aims to analyze the NC ratio of female and male pronuclei, and also to compare the size of single pronuclei using photographs that were obtained during experiments to create chimeric hermaphrodites from 2-cell oocytes. The volume of both the female and the male pronucleus was found to correlate with the volume of the oocyte cytoplasm. The NC ratio of the male pronucleus was greater than that of the female pronucleus. The NC ratio of the average volume of the female and male pronuclei was greater than the NC ratio of the mononucleate oocytes. The occurrence of 1PN oocytes was significantly higher when the volume of cytoplasm was lower than the cut-off value. These results indicated that the NC ratio is retained during pronuclear formation. A higher NC ratio in male compared with the female pronucleus indicated structural and/or molecular difference between the two pronuclei. 1PN formation may occur when sperm enters close to the MII spindle.
Collapse
|
5
|
Gambini A, Stein P, Savy V, Grow EJ, Papas BN, Zhang Y, Kenan AC, Padilla-Banks E, Cairns BR, Williams CJ. Developmentally Programmed Tankyrase Activity Upregulates β-Catenin and Licenses Progression of Embryonic Genome Activation. Dev Cell 2020; 53:545-560.e7. [PMID: 32442396 DOI: 10.1016/j.devcel.2020.04.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/16/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
Embryonic genome activation (EGA) is orchestrated by an intrinsic developmental program initiated during oocyte maturation with translation of stored maternal mRNAs. Here, we show that tankyrase, a poly(ADP-ribosyl) polymerase that regulates β-catenin levels, undergoes programmed translation during oocyte maturation and serves an essential role in mouse EGA. Newly translated TNKS triggers proteasomal degradation of axin, reducing targeted destruction of β-catenin and promoting β-catenin-mediated transcription of target genes, including Myc. MYC mediates ribosomal RNA transcription in 2-cell embryos, supporting global protein synthesis. Suppression of tankyrase activity using knockdown or chemical inhibition causes loss of nuclear β-catenin and global reductions in transcription and histone H3 acetylation. Chromatin and transcriptional profiling indicate that development arrests prior to the mid-2-cell stage, mediated in part by reductions in β-catenin and MYC. These findings indicate that post-transcriptional regulation of tankyrase serves as a ligand-independent developmental mechanism for post-translational β-catenin activation and is required to complete EGA.
Collapse
Affiliation(s)
- Andrés Gambini
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Paula Stein
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Virginia Savy
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Edward J Grow
- Department of Oncological Sciences, Huntsman Cancer Institute and Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Brian N Papas
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Yingpei Zhang
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Anna C Kenan
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Elizabeth Padilla-Banks
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Bradley R Cairns
- Department of Oncological Sciences, Huntsman Cancer Institute and Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Carmen J Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
6
|
Active Fluctuations of the Nuclear Envelope Shape the Transcriptional Dynamics in Oocytes. Dev Cell 2019; 51:145-157.e10. [DOI: 10.1016/j.devcel.2019.09.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/14/2019] [Accepted: 09/13/2019] [Indexed: 01/03/2023]
|
7
|
Schultz RM, Stein P, Svoboda P. The oocyte-to-embryo transition in mouse: past, present, and future. Biol Reprod 2019; 99:160-174. [PMID: 29462259 DOI: 10.1093/biolre/ioy013] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/03/2018] [Indexed: 02/06/2023] Open
Abstract
The oocyte-to-embryo transition (OET) arguably initiates with formation of a primordial follicle and culminates with reprogramming of gene expression during the course of zygotic genome activation. This transition results in converting a highly differentiated cell, i.e. oocyte, to undifferentiated cells, i.e. initial blastomeres of a preimplantation embryo. A plethora of changes occur during the OET and include, but are not limited to, changes in transcription, chromatin structure, and protein synthesis; accumulation of macromolecules and organelles that will comprise the oocyte's maternal contribution to the early embryo; sequential acquisition of meiotic and developmental competence to name but a few. This review will focus on transcriptional and post-transcriptional changes that occur during OET in mouse because such changes are likely the major driving force for OET. We often take a historical and personal perspective, and highlight how advances in experimental methods often catalyzed conceptual advances in understanding the molecular bases for OET. We also point out questions that remain open and therefore represent topics of interest for future investigation.
Collapse
Affiliation(s)
- Richard M Schultz
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Anatomy, Physiology, Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Paula Stein
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Petr Svoboda
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
8
|
Miki Y, Tsukamoto S, Minami N. How does the promoter of an oocyte-specific gene function in male germ cells? J Reprod Dev 2018; 64:463-468. [PMID: 30197401 PMCID: PMC6305850 DOI: 10.1262/jrd.2018-060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Studying gene expression in germ cells is useful for elucidating mechanisms of transcriptional regulation, because different genes are activated in male and female germ cells. The promoter regions of an oocyte-specific gene, Oog1, have been characterized. Driving the expression of green fluorescent protein with these different promoter regions provided us with critical information on the regulation of gene expression. The 3.9 kb long promoter functions in both male and female germ cells in transgenic mice. What is the cause of this sexually dimorphic expression? There may be important factors within and perhaps also outside this 3.9 kb promoter region that are required to maintain proper sex-specific gene expression.
Collapse
Affiliation(s)
- Yuka Miki
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Satoshi Tsukamoto
- Laboratory Animal and Genome Sciences Section, National Institute for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Naojiro Minami
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
9
|
Ladstätter S, Tachibana K. Genomic insights into chromatin reprogramming to totipotency in embryos. J Cell Biol 2018; 218:70-82. [PMID: 30257850 PMCID: PMC6314560 DOI: 10.1083/jcb.201807044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 12/19/2022] Open
Abstract
Ladstätter and Tachibana discuss changes in DNA methylation, chromatin accessibility, and topological architecture occurring during the reprogramming to totipotency in the early embryo. The early embryo is the natural prototype for the acquisition of totipotency, which is the potential of a cell to produce a whole organism. Generation of a totipotent embryo involves chromatin reorganization and epigenetic reprogramming that alter DNA and histone modifications. Understanding embryonic chromatin architecture and how this is related to the epigenome and transcriptome will provide invaluable insights into cell fate decisions. Recently emerging low-input genomic assays allow the exploration of regulatory networks in the sparsely available mammalian embryo. Thus, the field of developmental biology is transitioning from microscopy to genome-wide chromatin descriptions. Ultimately, the prototype becomes a unique model for studying fundamental principles of development, epigenetic reprogramming, and cellular plasticity. In this review, we discuss chromatin reprogramming in the early mouse embryo, focusing on DNA methylation, chromatin accessibility, and higher-order chromatin structure.
Collapse
Affiliation(s)
- Sabrina Ladstätter
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - Kikuë Tachibana
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| |
Collapse
|
10
|
Li J, Tang JX, Cheng JM, Hu B, Wang YQ, Aalia B, Li XY, Jin C, Wang XX, Deng SL, Zhang Y, Chen SR, Qian WP, Sun QY, Huang XX, Liu YX. Cyclin B2 can compensate for Cyclin B1 in oocyte meiosis I. J Cell Biol 2018; 217:3901-3911. [PMID: 30097513 PMCID: PMC6219713 DOI: 10.1083/jcb.201802077] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/12/2018] [Accepted: 08/01/2018] [Indexed: 11/22/2022] Open
Abstract
Cyclin B1 and its interaction with CDK1 are thought to be critical for meiosis I progression in oocytes. However, using oocyte-specific conditional knockouts, Li et al. show that Cyclin B2 activity can compensate for Cyclin B1 to trigger meiosis resumption. Mammalian oocytes are arrested at the prophase of the first meiotic division for months and even years, depending on species. Meiotic resumption of fully grown oocytes requires activation of M-phase–promoting factor (MPF), which is composed of Cyclin B1 and cyclin-dependent kinase 1 (CDK1). It has long been believed that Cyclin B1 synthesis/accumulation and its interaction with CDK1 is a prerequisite for MPF activation in oocytes. In this study, we revealed that oocyte meiotic resumption occurred in the absence of Cyclin B1. Ccnb1-null oocytes resumed meiosis and extruded the first polar body. Without Cyclin B1, CDK1 could be activated by up-regulated Cyclin B2. Ccnb1 and Ccnb2 double knockout permanently arrested the oocytes at the prophase of the first meiotic division. Oocyte-specific Ccnb1-null female mice were infertile due to failed MPF activity elevation and thus premature interphase-like stage entry in the second meiotic division. These results have revealed a hidden compensatory mechanism between Cyclin B1 and Cyclin B2 in regulating MPF and oocyte meiotic resumption.
Collapse
Affiliation(s)
- Jian Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Ji-Xin Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jin-Mei Cheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Bian Hu
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China.,Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Yu-Qian Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Batool Aalia
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiao-Yu Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Jin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiu-Xia Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Shou-Long Deng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yan Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Su-Ren Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei-Ping Qian
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Qing-Yuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xing-Xu Huang
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China.,Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Yi-Xun Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China .,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
11
|
Fraser R, Lin CJ. Epigenetic reprogramming of the zygote in mice and men: on your marks, get set, go! Reproduction 2016; 152:R211-R222. [PMID: 27601712 PMCID: PMC5097126 DOI: 10.1530/rep-16-0376] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/02/2016] [Indexed: 12/19/2022]
Abstract
Gametogenesis (spermatogenesis and oogenesis) is accompanied by the acquisition of gender-specific epigenetic marks, such as DNA methylation, histone modifications and regulation by small RNAs, to form highly differentiated, but transcriptionally silent cell-types in preparation for fertilisation. Upon fertilisation, extensive global epigenetic reprogramming takes place to remove the previously acquired epigenetic marks and produce totipotent zygotic states. It is the aim of this review to delineate the cellular and molecular events involved in maternal, paternal and zygotic epigenetic reprogramming from the time of gametogenesis, through fertilisation, to the initiation of zygotic genome activation for preimplantation embryonic development. Recent studies have begun to uncover the indispensable functions of epigenetic players during gametogenesis, fertilisation and preimplantation embryo development, and a more comprehensive understanding of these early events will be informative for increasing pregnancy success rates, adding particular value to assisted fertility programmes.
Collapse
Affiliation(s)
- Rupsha Fraser
- The University of EdinburghMRC Centre for Reproductive Health, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Chih-Jen Lin
- The University of EdinburghMRC Centre for Reproductive Health, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| |
Collapse
|
12
|
Lim CY, Knowles BB, Solter D, Messerschmidt DM. Epigenetic Control of Early Mouse Development. Curr Top Dev Biol 2016; 120:311-60. [PMID: 27475856 DOI: 10.1016/bs.ctdb.2016.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although the genes sequentially transcribed in the mammalian embryo prior to implantation have been identified, understanding of the molecular processes ensuring this transcription is still in development. The genomes of the sperm and egg are hypermethylated, hence transcriptionally silent. Their union, in the prepared environment of the egg, initiates their epigenetic genomic reprogramming into a totipotent zygote, in which the genome gradually becomes transcriptionally activated. During gametogenesis, sex-specific processes result in sperm and eggs with disparate epigenomes, both of which require drastic reprogramming to establish the totipotent genome of the zygote and the pluripotent inner cell mass of the blastocyst. Herein, we describe the factors, DNA and histone modifications, activation and repression of retrotransposons, and cytoplasmic localizations, known to influence the activation of the mammalian genome at the initiation of new life.
Collapse
Affiliation(s)
- C Y Lim
- Institute of Medical Biology, A*STAR, Singapore, Singapore
| | - B B Knowles
- Emerita, The Jackson Laboratory, Bar Harbor, ME, United States; Siriraj Center of Excellence for Stem Cell Research, Mahidol University, Bangkok, Thailand
| | - D Solter
- Siriraj Center of Excellence for Stem Cell Research, Mahidol University, Bangkok, Thailand; Emeritus, Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
| | - D M Messerschmidt
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore.
| |
Collapse
|
13
|
El-Hayek S, Clarke HJ. Control of Oocyte Growth and Development by Intercellular Communication Within the Follicular Niche. Results Probl Cell Differ 2016; 58:191-224. [PMID: 27300180 DOI: 10.1007/978-3-319-31973-5_8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In the mammalian ovary, each oocyte grows and develops within its own structural and developmental niche-the follicle. Together with the female germ cell in the follicle are somatic granulosa cells, specialized companion cells that surround the oocyte and provide support to it, and an outer layer of thecal cells that serve crucial roles including steroid synthesis. These follicular compartments function as a single physiological unit whose purpose is to produce a healthy egg, which upon ovulation can be fertilized and give rise to a healthy embryo, thus enabling the female germ cell to fulfill its reproductive potential. Beginning from the initial stage of follicle formation and until terminal differentiation at ovulation, oocyte and follicle growth depend absolutely on cooperation between the different cellular compartments. This cooperation synchronizes the initiation of oocyte growth with follicle activation. During growth, it enables metabolic support for the follicle-enclosed oocyte and allows the follicle to fulfill its steroidogenic potential. Near the end of the growth period, intra-follicular interactions prevent the precocious meiotic resumption of the oocyte and ensure its nuclear differentiation. Finally, cooperation enables the events of ovulation, including meiotic maturation of the oocyte and expansion of the cumulus granulosa cells. In this chapter, we discuss the cellular interactions that enable the growing follicle to produce a healthy oocyte, focusing on the communication between the germ cell and the surrounding granulosa cells.
Collapse
Affiliation(s)
- Stephany El-Hayek
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
- Department of Biology, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Block E-M0.2218, Montreal, QC, Canada, H4A 3J1
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada.
- Department of Biology, McGill University, Montreal, QC, Canada.
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Block E-M0.2218, Montreal, QC, Canada, H4A 3J1.
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
14
|
Su HJ, Zhang Y, Zhang L, Ma JL, Li JY, Pan KF, You WC. Methylation status of COX-2 in blood leukocyte DNA and risk of gastric cancer in a high-risk Chinese population. BMC Cancer 2015; 15:979. [PMID: 26674784 PMCID: PMC4682260 DOI: 10.1186/s12885-015-1962-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 11/30/2015] [Indexed: 01/27/2023] Open
Abstract
Background Methylation is a common epigenetic modification which may play a crucial role in cancer development. To investigate the association between methylation of COX-2 in blood leukocyte DNA and risk of gastric cancer (GC), a nested case–control study was conducted in Linqu County, Shandong Province, a high risk area of GC in China. Methods Association between blood leukocyte DNA methylation of COX-2 and risk of GC was investigated in 133 GCs and 285 superficial gastritis (SG)/ chronic atrophic gastritis (CAG). The temporal trend of COX-2 methylation level during GC development was further explored in 74 pre-GC and 95 post-GC samples (including 31 cases with both pre- and post-GC samples). In addition, the association of DNA methylation and risk of progression to GC was evaluated in 74 pre-GC samples and their relevant intestinal metaplasia (IM)/dysplasia (DYS) controls. Methylation level was determined by quantitative methylation-specific PCR (QMSP). Odds ratios (ORs) and 95 % confidence intervals (CIs) were calculated by unconditional logistic regression analysis. Results The medians of COX-2 methylation levels were 2.3 % and 2.2 % in GC cases and controls, respectively. No significant association was found between COX-2 methylation and risk of GC (OR, 1.15; 95 % CI: 0.70-1.88). However, the temporal trend analysis showed that COX-2 methylation levels were elevated at 1–4 years ahead of clinical GC diagnosis compared with the year of GC diagnosis (3.0 % vs. 2.2 %, p = 0.01). Further validation in 31 GCs with both pre- and post-GC samples indicated that COX-2 methylation levels were significantly decreased at the year of GC diagnosis compared with pre-GC samples (1.5 % vs. 2.5 %, p = 0.02). No significant association between COX-2 methylation and risk of progression to GC was found in subjects with IM (OR, 0.50; 95 % CI: 0.18–1.42) or DYS (OR, 0.70; 95 % CI: 0.23–2.18). Additionally, we found that elder people had increased risk of COX-2 hypermethylation (OR, 1.55; 95 % CI: 1.02–2.36) and subjects who ever infected with H. pylori had decreased risk of COX-2 hypermethylation (OR, 0.54; 95 % CI: 0.34–0.88). Conclusions COX-2 methylation exists in blood leukocyte DNA but at a low level. COX-2 methylation levels in blood leukocyte DNA may change during GC development.
Collapse
Affiliation(s)
- Hui-juan Su
- Key Laboratory of Carcinogenesis and Translation Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| | - Yang Zhang
- Key Laboratory of Carcinogenesis and Translation Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| | - Lian Zhang
- Key Laboratory of Carcinogenesis and Translation Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| | - Jun-ling Ma
- Key Laboratory of Carcinogenesis and Translation Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| | - Ji-You Li
- Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| | - Kai-feng Pan
- Key Laboratory of Carcinogenesis and Translation Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| | - Wei-cheng You
- Key Laboratory of Carcinogenesis and Translation Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| |
Collapse
|
15
|
Yamamoto R, Abe KI, Suzuki Y, Suzuki MG, Aoki F. Characterization of gene expression in mouse embryos at the 1-cell stage. J Reprod Dev 2015; 62:87-92. [PMID: 26599803 PMCID: PMC4768782 DOI: 10.1262/jrd.2015-131] [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] [Indexed: 11/20/2022] Open
Abstract
In mice, transcription from the zygotic genome is initiated at the mid-1-cell stage after fertilization.
Although a recent high-throughput sequencing (HTS) analysis revealed that this transcription occurs
promiscuously throughout almost the entire genome in 1-cell stage embryos, a detailed investigation of this
process has yet to be conducted using protein-coding genes. Thus, the present study utilized previous RNA
sequencing (RNAseq) data to determine the characteristics and regulatory regions of genes transcribed at the
1-cell stage. While the expression patterns of protein-coding genes of mouse embryos were very different at
the 1-cell stage than at other stages and in various tissues, an analysis for the upstream and downstream
regions of actively expressed genes did not reveal any elements that were specific to 1-cell stage embryos.
Therefore, the unique gene expression pattern observed at the 1-cell stage in mouse embryos appears to be
governed by mechanisms independent of a specific promoter element.
Collapse
Affiliation(s)
- Ryoma Yamamoto
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Chiba 277-8562, Japan
| | | | | | | | | |
Collapse
|
16
|
Xia X, Yan C, Wu W, Zhou Y, Hou L, Zuo B, Xu D, Ren Z, Xiong Y. Characterization of the porcine peptidylarginine deiminase type VI gene (PADI6) promoter: Sp1 regulates basal transcription of the porcine PADI6. Gene 2015; 575:551-558. [PMID: 26403316 DOI: 10.1016/j.gene.2015.09.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 09/13/2015] [Accepted: 09/16/2015] [Indexed: 11/19/2022]
Abstract
It is a general consensus that oocyte quality is the key to embryo survival in pig reproduction. Thus, study on regulation of the ovary-associated gene is of great significance in pig breeding. Peptidylarginine deiminases (PADs) are a family of enzymes which catalyze the conversion of arginine to citrulline in proteins. The peptidylarginine deiminases type VI gene (PADI6) is mainly expressed in the ovary, and plays an important role in oocyte growth, fertilization and early embryo development. However, until now, little is known about its transcriptional regulation mechanism. Here, we firstly isolated and characterized the 5'-flanking region of porcine PADI6 gene. We determined the transcription start site using 5'-rapid amplification of cDNA ends (RACE) analysis, and identified the minimal promoter (-85/+68) that drove the basal expression of PADI6 by constructing various progressive deletions. Mutational analysis and electrophoretic mobility shift assays demonstrated Sp1 bound to the -56/-47 region of the PADI6 promoter. Furthermore, overexpression of Sp1 significantly increased the promoter activity and promoted PADI6 gene expression, and accordingly, inhibition of Sp1 expression with specific siRNA significantly reduced the promoter activity and suppressed the PADI6 expression. In addition, inhibition of Sp1 binding by Mithramycin A treatment reduced the transcriptional activity of PADI6 in a dose-dependent manner. Taken together, these data indicate that Sp1 is essential for the transcriptional regulation of PADI6.
Collapse
Affiliation(s)
- Xiaoliang Xia
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Sciences, Huazhong Agricultural University, Wuhan 430070, China.
| | - Chi Yan
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Sciences, Huazhong Agricultural University, Wuhan 430070, China.
| | - Wangjun Wu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Ying Zhou
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Sciences, Huazhong Agricultural University, Wuhan 430070, China.
| | - Liming Hou
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Sciences, Huazhong Agricultural University, Wuhan 430070, China.
| | - Bo Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Sciences, Huazhong Agricultural University, Wuhan 430070, China.
| | - Dequan Xu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Sciences, Huazhong Agricultural University, Wuhan 430070, China.
| | - Zhuqing Ren
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Sciences, Huazhong Agricultural University, Wuhan 430070, China.
| | - Yuanzhu Xiong
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Sciences, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
17
|
Labrecque R, Lodde V, Dieci C, Tessaro I, Luciano AM, Sirard MA. Chromatin remodelling and histone m RNA accumulation in bovine germinal vesicle oocytes. Mol Reprod Dev 2015; 82:450-62. [PMID: 25940597 DOI: 10.1002/mrd.22494] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/14/2015] [Indexed: 01/24/2023]
Abstract
Major remodelling of the chromatin enclosed within the germinal vesicle occurs towards the end of oocyte growth in mammals, but the mechanisms involved in this process are not completely understood. In bovine, four distinct stages of chromatin compaction-ranging from a diffused state (GV0) to a fully compacted configuration (GV3)-are linked to the gradual acquisition of developmental potential. To better understand the molecular events and to identify mRNA modulations occurring in the oocyte during the GV0-to-GV3 transition, transcriptomic analysis was performed with the EmbryoGENE microarray platform. The mRNA abundance of several genes decreased as chromatin compaction increased, which correlates with progressive transcriptional silencing that is characteristic of the end of oocyte growth. On the other hand, the abundance of some transcripts increased during the same period, particularly several histone gene transcripts from the H2A, H2B, H3, H4, and linker H1 family. In silico analysis predicted RNA-protein interactions between specific histone transcripts and the bovine stem-loop binding protein 2 (SLBP2), which helps regulate the translation of histone mRNA during oogenesis. These results suggest that some histone-encoding transcripts are actively stored, possibly to sustain the needs of the embryo before genome activation. This dataset offers a unique opportunity to survey which histone mRNAs are needed to complete chromatin compaction during oocyte maturation and which are stockpiled for the first three cell cycles following fertilization.
Collapse
Affiliation(s)
- R Labrecque
- Département des Sciences Animales, Centre de Recherche en Biologie de la Reproduction, Université Laval, Québec, Québec, Canada
| | - V Lodde
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Sciences and Food Safety, University of Milan, Milan, Italy
| | - C Dieci
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Sciences and Food Safety, University of Milan, Milan, Italy
| | - I Tessaro
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Sciences and Food Safety, University of Milan, Milan, Italy
| | - A M Luciano
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Sciences and Food Safety, University of Milan, Milan, Italy
| | - M A Sirard
- Département des Sciences Animales, Centre de Recherche en Biologie de la Reproduction, Université Laval, Québec, Québec, Canada
- Department of Animal Sciences, Laval University, Québec, Québec, Canada
| |
Collapse
|
18
|
Diao YF, Oqani RK, Li XX, Lin T, Kang JW, Jin DI. Changes in histone H3 lysine 36 methylation in porcine oocytes and preimplantation embryos. PLoS One 2014; 9:e100205. [PMID: 24927323 PMCID: PMC4057445 DOI: 10.1371/journal.pone.0100205] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 05/23/2014] [Indexed: 01/31/2023] Open
Abstract
Histone H3 lysine 36 (H3K36) methylation is known to be associated with transcriptionally active genes, and is considered a genomic marker of active loci. To investigate the changes in H3K36 methylation in pig, we determined the mono-, di-, and tri-methylations of H3K36 (H3K36me1, H3K36me2 and H3K36me3, respectively) in porcine fetal fibroblasts, oocytes and preimplantation embryos by immunocytochemistry using specific antibodies and confocal microscopy. These analyses revealed that only H3K36me3 in porcine fetal fibroblasts consistently colocalized with transcription sites identified as actively synthesizing RNA based on fluorouridine (FU) incorporation. Treatment of cells with flavopiridol, which blocks transcription elongation, completely abrogated both H3K36me3 signals and RNA synthesis. All three types of H3K36 methylation were present and did not significantly differ during oocyte maturation. In parthenogenetic embryos, H3K36me1 and -me2 were detected in 1-cell through blastocyst-stage embryos. In contrast, H3K36me3 was not detected in most 1-cell stage embryos. H3K36me3 signals became detectable in 2-cell stage embryos, peaked at the 4-cell stage, decreased at the 8-cell stage, and then became undetectable at blastocyst stages in both parthenogenetic and in vitro-fertilized (IVF) embryos. Unlike the case in IVF embryos, H3K36me3 could not be demethylated completely during the 1-cell stage in somatic cell nuclear transfer (SCNT) embryos. These results collectively indicate that H3K36me3, but not H3K36me1 or -me2, is associated with transcription elongation in porcine fetal fibroblasts. H3K36me3 is developmentally regulated and may be a histone mark of embryonic gene activation in pig. Aberrant H3K36 tri-methylation occurred during the nuclear reprogramming of SCNT embryos.
Collapse
Affiliation(s)
- Yun Fei Diao
- Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pigs, Chungnam National University, Daejeon, Korea
| | - Reza K Oqani
- Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pigs, Chungnam National University, Daejeon, Korea
| | - Xiao Xia Li
- Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pigs, Chungnam National University, Daejeon, Korea
| | - Tao Lin
- Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pigs, Chungnam National University, Daejeon, Korea
| | - Jung Won Kang
- Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pigs, Chungnam National University, Daejeon, Korea
| | - Dong Il Jin
- Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pigs, Chungnam National University, Daejeon, Korea
| |
Collapse
|
19
|
Nuclear distribution of RNA polymerase II and mRNA processing machinery in early mammalian embryos. BIOMED RESEARCH INTERNATIONAL 2014; 2014:681596. [PMID: 24868542 PMCID: PMC4020508 DOI: 10.1155/2014/681596] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 04/11/2014] [Indexed: 11/17/2022]
Abstract
Spatial distribution of components of nuclear metabolism provides a significant impact on regulation of the processes of gene expression. While distribution of the key nuclear antigens and their association with the defined nuclear domains were thoroughly traced in mammalian somatic cells, similar data for the preimplantation embryos are scanty and fragmental. However, the period of cleavage is characterized by the most drastic and dynamic nuclear reorganizations accompanying zygotic gene activation. In this minireview, we try to summarize the results of studies concerning distribution of major factors involved in RNA polymerase II-dependent transcription, pre-mRNA splicing mRNA export that have been carried out on early embryos of mammals.
Collapse
|
20
|
Park SJ, Komata M, Inoue F, Yamada K, Nakai K, Ohsugi M, Shirahige K. Inferring the choreography of parental genomes during fertilization from ultralarge-scale whole-transcriptome analysis. Genes Dev 2014; 27:2736-48. [PMID: 24352427 PMCID: PMC3877761 DOI: 10.1101/gad.227926.113] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The mechanisms underlying parental genome coordination upon mammalian fertilization remain elusive due to difficulties in preparing large numbers of high-quality preimplantation cells. Here, Park et al. collect an unprecedented number of mouse oocytes and establish detailed transcriptional profiles for four early embryonic stages and parthenogenetic development. Bioinformatic analysis identifies a distinctive gene regulatory network activated in embryos after fertilization compared with parthenotes. This large-scale profile of early mouse embryos yields a valuable resource for developmental biology and stem cell research. Fertilization precisely choreographs parental genomes by using gamete-derived cellular factors and activating genome regulatory programs. However, the mechanism remains elusive owing to the technical difficulties of preparing large numbers of high-quality preimplantation cells. Here, we collected >14 × 104 high-quality mouse metaphase II oocytes and used these to establish detailed transcriptional profiles for four early embryo stages and parthenogenetic development. By combining these profiles with other public resources, we found evidence that gene silencing appeared to be mediated in part by noncoding RNAs and that this was a prerequisite for post-fertilization development. Notably, we identified 817 genes that were differentially expressed in embryos after fertilization compared with parthenotes. The regulation of these genes was distinctly different from those expressed in parthenotes, suggesting functional specialization of particular transcription factors prior to first cell cleavage. We identified five transcription factors that were potentially necessary for developmental progression: Foxd1, Nkx2-5, Sox18, Myod1, and Runx1. Our very large-scale whole-transcriptome profile of early mouse embryos yielded a novel and valuable resource for studies in developmental biology and stem cell research. The database is available at http://dbtmee.hgc.jp.
Collapse
Affiliation(s)
- Sung-Joon Park
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo 108-8639, Japan
| | | | | | | | | | | | | |
Collapse
|
21
|
Hamamoto G, Suzuki T, Suzuki MG, Aoki F. Regulation of transketolase like 1 gene expression in the murine one-cell stage embryos. PLoS One 2014; 9:e82087. [PMID: 24392079 PMCID: PMC3879240 DOI: 10.1371/journal.pone.0082087] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 10/18/2013] [Indexed: 12/03/2022] Open
Abstract
In mice, transcription from the zygotic genome starts at the mid-one-cell stage after fertilization. Previous studies showed that an enhancer is not required for transcription at this stage, and that the enhancer-dependent mechanism of transcription is established during the two-cell stage. However, these results were obtained using reporter gene assays with promoters derived from viruses, rather than from endogenous genes. We conducted a reporter-gene assay using the promoter of Tktl1, which is transcribed after fertilization, to investigate the mechanism regulating gene expression at the one-cell stage. When a plasmid containing the 2467 bp upstream and 25 bp downstream of the Tktl1 transcription start site (TSS) was microinjected into the nuclei of growing oocytes, and one-cell stage and early and late two-cell-stage embryos, transcriptional activity was detected in the one-cell- and two-cell-stage embryos, but not in the oocytes. It was highest at the early two-cell stage and was reduced at the late two-cell stage. The decrease in activity at the late two-cell stage was prevented by inhibiting the second round of DNA replication, suggesting that the transcriptionally repressive state is established during the two-cell stage by a mechanism coupled to DNA replication. When the Tktl1 promoter was deleted to leave 56 bp upstream of the TSS which includes GC and TATA boxes, transcriptional activity was still detected in one-cell-stage embryos, but not early or late two-cell-stage embryos. The core promoter of Tktl1 alone seems to be able to induce basal transcription at the one-cell stage. These results suggest that repressive chromatin is established after fertilization in two steps, which occur during the transition from the one- to two-cell stage and during DNA replication at the two-cell stage.
Collapse
Affiliation(s)
- Go Hamamoto
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Chiba, Japan
| | - Tsukasa Suzuki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Chiba, Japan
| | - Masataka G. Suzuki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Chiba, Japan
| | - Fugaku Aoki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Chiba, Japan
- * E-mail:
| |
Collapse
|
22
|
Castillo J, Amaral A, Oliva R. Sperm nuclear proteome and its epigenetic potential. Andrology 2013; 2:326-38. [PMID: 24327354 DOI: 10.1111/j.2047-2927.2013.00170.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/08/2013] [Accepted: 11/11/2013] [Indexed: 11/29/2022]
Abstract
The main function of the sperm cell is to transmit the paternal genetic message and epigenetic information to the embryo. Importantly, the majority of the genes in the sperm chromatin are highly condensed by protamines, whereas genes potentially needed in the initial stages of development are associated with histones, representing a form of epigenetic marking. However, so far little attention has been devoted to other sperm chromatin-associated proteins that, in addition to histones and protamines, may also have an epigenetic role. Therefore, with the goal of contributing to cover this subject we have compiled, reviewed and report a list of 581 chromatin or nuclear proteins described in the human sperm cell. Furthermore, we have analysed their Gene Ontology Biological Process enriched terms and have grouped them into different functional categories. Remarkably, we show that 56% of the sperm nuclear proteins have a potential epigenetic activity, being involved in at least one of the following functions: chromosome organization, chromatin organization, protein-DNA complex assembly, DNA packaging, gene expression, transcription, chromatin modification and histone modification. In addition, we have also included and compared the sperm cell proteomes of different model species, demonstrating the existence of common trends in the chromatin composition in the mammalian mature male gamete. Taken together, our analyses suggest that the mammalian sperm cell delivers to the offspring a rich combination of histone variants, transcription factors, chromatin-associated and chromatin-modifying proteins which have the potential to encode and transmit an extremely complex epigenetic information.
Collapse
Affiliation(s)
- J Castillo
- Human Genetics Research Group, IDIBAPS, Faculty of Medicine, University of Barcelona, Barcelona, Spain; Biochemistry and Molecular Genetics Service, Hospital Clinic, Barcelona, Spain
| | | | | |
Collapse
|
23
|
Ishida M, Okazaki E, Tsukamoto S, Kimura K, Aizawa A, Kito S, Imai H, Minami N. The promoter of the oocyte-specific gene, Oog1, functions in both male and female meiotic germ cells in transgenic mice. PLoS One 2013; 8:e68686. [PMID: 23894331 PMCID: PMC3718783 DOI: 10.1371/journal.pone.0068686] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Accepted: 06/02/2013] [Indexed: 12/05/2022] Open
Abstract
Oog1 is an oocyte-specific gene whose expression is turned on in mouse oocytes at embryonic day (E) 15.5, concomitant with the time when most of the female germ cells stop proliferating and enter meiotic prophase. Here, we characterize the Oog1 promoter, and show that transgenic GFP reporter expression driven by the 2.7 kb and 3.9 kb regions upstream of the Oog1 transcription start site recapitulates the intrinsic Oog1 expression pattern. In addition, the 3.9 kb upstream region exhibits stronger transcriptional activity than does the 2.7 kb region, suggesting that regulatory functions might be conserved in the additional 1.2 kb region found within the 3.9 kb promoter. Interestingly, the longer promoter (3.9 kb) also showed strong activity in male germ cells, from late pachytene spermatocytes to elongated spermatids. This is likely due to the aberrant demethylation of two CpG sites in the proximal promoter region. One was highly methylated in the tissues in which GFP expression was suppressed, and another was completely demethylated only in Oog1pro3.9 male and female germ cells. These results suggest that aberrant demethylation of the proximal promoter region induced ectopic expression in male germ cells under the control of 3.9 kb Oog1 promoter. This is the first report indicating that sex-dependent gene expression is altered according to the length and the methylation status of the promoter region. Additionally, our results show that individual CpG sites are differentially methylated and play different roles in regulating promoter activity and gene transcription.
Collapse
Affiliation(s)
- Miya Ishida
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Eriko Okazaki
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Satoshi Tsukamoto
- Laboratory of Animal and Genome Science Section, National Institute of Radiological Sciences, Chiba, Japan
| | - Koji Kimura
- Animal Reproduction Laboratory, National Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Nasushiobara, Japan
| | | | - Seiji Kito
- Laboratory of Animal and Genome Science Section, National Institute of Radiological Sciences, Chiba, Japan
| | - Hiroshi Imai
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Naojiro Minami
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
24
|
Sun SC, Wang XG, Ma XS, Huang XJ, Li J, Liu HL. TBP dynamics during mouse oocyte meiotic maturation and early embryo development. PLoS One 2013; 8:e55425. [PMID: 23383188 PMCID: PMC3561223 DOI: 10.1371/journal.pone.0055425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 12/22/2012] [Indexed: 11/18/2022] Open
Abstract
To maintain cell lineage, cells develop a mechanism which can transmit the gene activity information to the daughter cells. In mitosis, TBP (TATA-binding protein), a transcription factor which belongs to TFIID was associated with M phase chromosomes and was proved to be a bookmark for cellular memory. Although previous work showed that TBP was dispensable for mouse oocyte maturation and early embryo development, exogenous TBP protein was detected in the nuclear of oocytes and early embryos. It is still unknown whether exogenous TBP can associate with condensed chromosomes during meiosis and mouse early embryo development. In present study by the injection of GFP-tagged TBP mRNA we for the first time investigated TBP dynamics in mouse early embryos and confirmed its localization pattern in oocytes. The exogenous TBP enriched at germinal vesicle at GV stage but disappeared from the chromosomes after GVBD. Moreover, exogenous TBP was still dispersed from the chromosomes of somatic donor nuclear in oocytes by nuclear transfer (NT), further proving that oocyte has some mechanism to remove TBP. During mouse embryo development, the exogenous TBP was removed from the chromosomes of M phase zygotes, but was found to express weakly at the M phase of 2-cell. Moreover, in the blastocyst TBP was also detected at the M phase chromosomes. Overexpression of TBP caused the failure of oocyte maturation and embryo development. Our results supported the idea that TBP might be a marker for transmitting cellular memory to daughter cells.
Collapse
Affiliation(s)
- Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xu-Guang Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xue-Shan Ma
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xian-Ju Huang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Juan Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Hong-Lin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- * E-mail:
| |
Collapse
|
25
|
Climent M, Alonso-Martin S, Pérez-Palacios R, Guallar D, Benito AA, Larraga A, Fernández-Juan M, Sanz M, de Diego A, Seisdedos MT, Muniesa P, Schoorlemmer J. Functional analysis of Rex1 during preimplantation development. Stem Cells Dev 2012; 22:459-72. [PMID: 22897771 DOI: 10.1089/scd.2012.0211] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Rex1/Zfp42 is a nuclear protein that is highly conserved in mammals, and widely used as an embryonic stem (ES) cell marker. Although Rex1 expression is associated with enhanced pluripotency, loss-of-function models recently described do not exhibit major phenotypes, and both preimplantation development and ES cell derivation appear normal in the absence of Rex1. To better understand the functional role of Rex1, we examined the expression and localization of Rex1 during preimplantation development. Our studies indicated that REX1 is expressed at all stages during mouse preimplantation development, with a mixed pattern of nuclear, perinuclear, and cytoplasmic localization. Chromatin association seemed to be altered in 8-cell embryos, and in the blastocyst, we found REX1 localized almost exclusively in the nucleus. A functional role for Rex1 in vivo was assessed by gain- and loss-of-function approaches. Embryos with attenuated levels of Rex1 after injection of zygotes with siRNAs did not exhibit defects in preimplantation development in vitro. In contrast, overexpression of Rex1 interfered with cleavage divisions and with proper blastocyst development, although we failed to detect alterations in the expression of lineage and pluripotency markers. Rex1 gain- and loss-of-function did alter the expression levels of Zscan4, an important regulator of preimplantation development and pluripotency. Our results suggest that Rex1 plays a role during preimplantation development. They are compatible with a role for Rex1 during acquisition of pluripotency in the blastocyst.
Collapse
Affiliation(s)
- María Climent
- Departamento de Anatomía, Embriología y Genética Animal, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Xia M, He H, Wang Y, Liu M, Zhou T, Lin M, Zhou Z, Huo R, Zhou Q, Sha J. PCBP1 is required for maintenance of the transcriptionally silent state in fully grown mouse oocytes. Cell Cycle 2012; 11:2833-42. [PMID: 22801551 DOI: 10.4161/cc.21169] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Global transcriptional silencing in fully grown oocytes is a critical event during mammalian oogenesis. However, how this event is regulated remains elusive. Here, we provide evidence that poly(rC)-binding protein 1 (PCBP1), a protein found by us previously to be present in metaphase II (MII) mouse oocytes, participates in maintenance of the transcriptionally silent state in fully grown mouse oocytes. Knocking down Pcbp1 by microinjection of its specific siRNAs into fully grown germinal vesicle (GV) oocytes resulted in remarkable changes in their transcriptional state, including the disequilibrium between the number of oocytes with an NSN (non-surrounded nucleolus) and those with a SN (surrounded nucleolus), and obvious transcriptional reactiviation in oocytes with a SN configuration as evidenced by BrUTP incorporation assay and immunofluorescent labeling of phosphorylated RNA polymerase II CTD and trimethylated H3 lysine 4, markers for active transcription. Furthermore, in a comprehensive microarray analysis of the preovulatory oocyte transcriptome, an incredible number of nearly 4,000 transcripts were upregulated in the Pcbp1 knockdown groups. These data indicate that lack of the function of PCBP1 disrupts the quiescent status of transcription in the fully grown oocytes, and hence supporting a role of this protein in the regulation of global transcriptional silcencing in fully grown mouse oocytes.
Collapse
Affiliation(s)
- Meng Xia
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yue MX, Fu XW, Zhou GB, Hou YP, DU M, Wang L, Zhu SE. Abnormal DNA methylation in oocytes could be associated with a decrease in reproductive potential in old mice. J Assist Reprod Genet 2012; 29:643-50. [PMID: 22618193 DOI: 10.1007/s10815-012-9780-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Accepted: 04/24/2012] [Indexed: 12/28/2022] Open
Abstract
PURPOSE This study was designed to evaluate DNA methylation and the expression of DNA methyltransferases (Dnmt1, Dnmt3a, Dnmt3b and Dnmt3L) in metaphaseII (MII) oocytes and the DNA methylation of pre-implantation embryos during mouse aging to address whether such aging-related changes are associated with decreased reproductive potential in aged mice. METHODS Oocytes (MII) from 6 to 8 weeks old female mice are referred to as the 'young group'; oocytes from the same group that were maintained until 35-40 weeks old are referred to as the 'old group.' The oocytes were fertilized both in vitro and in vivo to obtain embryos. The DNA methylation levels in the oocytes (MII) and pre-implantation embryos were assessed using fluorescence staining. The expression levels of the Dnmt genes in the oocytes (MII) were assessed using Western blotting. RESULTS The DNA methylation levels in the oocytes and pre-implantation embryos (in vivo and in vitro) decreased significantly during the aging of the mice. The expression levels of all of the examined Dnmt proteins in the old group were lower than young group. Both the cleavage and blastocyst rate were significantly lower in the oocytes of the older mice (69.9 % vs. 80.9 %, P < 0.05; 33.9 % vs. 56.4 %, P < 0.05). The pregnancy rate of the old mice was lower than that of the young mice (46.7 % vs. 100 %, P < 0.05). The stillbirth and fetal malformation rate was significantly higher in the old group than in the young group (17.2 % vs. 2.9 %, P < 0.05). CONCLUSIONS The decreased expression of Dnmt1, Dnmt3a, Dnmt3b and Dnmt3L in oocytes (MII) and the change of genome-wide DNA methylation in oocytes and pre-implantation embryos due to aging may be related to lower reproductive potential in old female mice.
Collapse
Affiliation(s)
- Ming-xing Yue
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
28
|
Vassena R, Boué S, González-Roca E, Aran B, Auer H, Veiga A, Izpisua Belmonte JC. Waves of early transcriptional activation and pluripotency program initiation during human preimplantation development. Development 2011; 138:3699-709. [PMID: 21775417 DOI: 10.1242/dev.064741] [Citation(s) in RCA: 204] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The events regulating human preimplantation development are still largely unknown owing to a scarcity of material, ethical and legal limitations and a lack of reliable techniques to faithfully amplify the transcriptome of a single cell. Nonetheless, human embryology is gathering renewed interest due to its close relationship with both stem cell biology and epigenetic reprogramming to pluripotency and their importance in regenerative medicine. Carefully timed genome-wide transcript analyses of single oocytes and embryos uncovered a series of successive waves of embryonic transcriptional initiation that start as early as the 2-cell stage. In addition, we identified the hierarchical activation of genes involved in the regulation of pluripotency. Finally, we developed HumER, a database of human preimplantation gene expression, to serve the scientific community. Importantly, our work links early transcription in the human embryo with the correct execution of the pluripotency program later in development and paves the way for the identification of factors to improve epigenetic reprogramming.
Collapse
Affiliation(s)
- Rita Vassena
- Center for Regenerative Medicine in Barcelona, CMR[B], Dr Aiguader 88, 08003, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
29
|
Johnson GD, Lalancette C, Linnemann AK, Leduc F, Boissonneault G, Krawetz SA. The sperm nucleus: chromatin, RNA, and the nuclear matrix. Reproduction 2011; 141:21-36. [PMID: 20876223 PMCID: PMC5358669 DOI: 10.1530/rep-10-0322] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Within the sperm nucleus, the paternal genome remains functionally inert and protected following protamination. This is marked by a structural morphogenesis that is heralded by a striking reduction in nuclear volume. Despite these changes, both human and mouse spermatozoa maintain low levels of nucleosomes that appear non-randomly distributed throughout the genome. These regions may be necessary for organizing higher order genomic structure through interactions with the nuclear matrix. The promoters of this transcriptionally quiescent genome are differentially marked by modified histones that may poise downstream epigenetic effects. This notion is supported by increasing evidence that the embryo inherits these differing levels of chromatin organization. In concert with the suite of RNAs retained in the mature sperm, they may synergistically interact to direct early embryonic gene expression. Irrespective, these features reflect the transcriptional history of spermatogenic differentiation. As such, they may soon be utilized as clinical markers of male fertility. In this review, we explore and discuss how this may be orchestrated.
Collapse
Affiliation(s)
- Graham D. Johnson
- The Center for Molecular Medicine and Genetics, Wayne State University of Medicine, C.S. Mott Center, 275 E. Hancock, Detroit, MI 48201
| | - Claudia Lalancette
- The Center for Molecular Medicine and Genetics, Wayne State University of Medicine, C.S. Mott Center, 275 E. Hancock, Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University of Medicine, C.S. Mott Center, 275 E. Hancock, Detroit, MI 48201
| | - Amelia K. Linnemann
- The Center for Molecular Medicine and Genetics, Wayne State University of Medicine, C.S. Mott Center, 275 E. Hancock, Detroit, MI 48201
| | - Frédéric Leduc
- Department of Biochemistry, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada J1H 5N4
| | - Guylain Boissonneault
- Department of Biochemistry, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada J1H 5N4
| | - Stephen A. Krawetz
- The Center for Molecular Medicine and Genetics, Wayne State University of Medicine, C.S. Mott Center, 275 E. Hancock, Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University of Medicine, C.S. Mott Center, 275 E. Hancock, Detroit, MI 48201
- Institute for Scientific Computing, Wayne State University of Medicine, C.S. Mott Center, 275 E. Hancock, Detroit, MI 48201
| |
Collapse
|
30
|
Steger K, Cavalcanti MCO, Schuppe HC. Prognostic markers for competent human spermatozoa: fertilizing capacity and contribution to the embryo. ACTA ACUST UNITED AC 2010; 34:513-27. [DOI: 10.1111/j.1365-2605.2010.01129.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
31
|
Probst AV, Okamoto I, Casanova M, El Marjou F, Le Baccon P, Almouzni G. A strand-specific burst in transcription of pericentric satellites is required for chromocenter formation and early mouse development. Dev Cell 2010; 19:625-38. [PMID: 20951352 DOI: 10.1016/j.devcel.2010.09.002] [Citation(s) in RCA: 246] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 07/01/2010] [Accepted: 08/18/2010] [Indexed: 01/21/2023]
Abstract
At the time of fertilization, the paternal genome lacks the typical configuration and marks characteristic of pericentric heterochromatin. It is thus essential to understand the dynamics of this region during early development, its importance during that time period and how a somatic configuration is attained. Here, we show that pericentric satellites undergo a transient peak in expression precisely at the time of chromocenter formation. This transcription is regulated in a strand-specific manner in time and space and is strongly biased by the parental asymmetry. The transcriptional upregulation follows a developmental clock, yet when replication is blocked chromocenter formation is impeded. Furthermore, interference with major satellite transcripts using locked nucleic acid (LNA)-DNA gapmers results in developmental arrest before completion of chromocenter formation. We conclude that the exquisite strand-specific expression dynamics at major satellites during the 2-cell stage, with both up and downregulation, are necessary events for proper chromocenter organization and developmental progression.
Collapse
Affiliation(s)
- Aline V Probst
- Laboratory of Nuclear Dynamics and Genome Plasticity, Unité Mixte de Recherche, 218 Centre National de la Recherche Scientifique/Institut Curie, 26, rue d'Ulm, 75248 Paris Cedex 05, France
| | | | | | | | | | | |
Collapse
|
32
|
Beaujean N, Mason K, Bonnet-Garnier A, Salvaing J, Debey P. [Embryonic genome organization after fertilization in mammals]. Biol Aujourdhui 2010; 204:205-13. [PMID: 20950564 DOI: 10.1051/jbio/2010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Indexed: 11/15/2022]
Abstract
In mammals, the embryonic genome is first transcriptionally inactive after fertilization. Embryonic development is then strictly dependent on the maternally inherited RNA and proteins accumulated before ovulation and present in the oocyte cytoplasm. The onset of embryonic gene expression is initiated later during development, i.e. during the "embryonic genome activation (EGA)". EGA takes place at various preimplantation stages according to species and is dependent on the presence of the basal transcriptional machinery components but also on parental genomes reorganizations after fertilization. Indeed, during the first embryonic cycles, nuclei undergo intense remodeling that could be a key regulator of embryonic development.
Collapse
Affiliation(s)
- Nathalie Beaujean
- INRA, UMR1198 Biologie du Développement et Reproduction, 78352 Jouy-en-Josas, France.
| | | | | | | | | |
Collapse
|
33
|
Abstract
Ejaculated mammalian spermatozoa contain a complex yet specific population of mRNA. However, the possible roles that mRNA has in early zygotic and embryonic development remain unclear. We found that Dby mRNA is selectively retained in capacitated mouse spermatozoa, and is transferred into the oocyte during fertilization by reverse transcription-polymerase chain reaction even though no DBY protein expression is detected. The cellular location of Dby mRNA is seen in the post-acrosome region, and it comprises nearly half of the mouse spermatozoa in in situ hybridization. In contrast, transcripts of the control gene, Smcy, are not detected in capacitated mouse spermatozoa, although the H-Y antigen encoded by Smcy is expressed on the surface of the spermatozoa. In our microinjection experiment, the zygotic development rate of the as-Dby male pronucleus injection group was significantly lower than that of the as-Smcy male pronucleus injection group (35.9% vs. 95%, P = 0.001) and the as-Dby female pronucleus injection group (35.9% vs. 93.8%, P = 0.001). The rate of male-developed zygotes was also lower than that of the as-Smcy male pronucleus injection group (17.4% vs. 57.9%, P = 0.002) and the as-Dby female pronucleus injection group (17.4% vs. 54.1%, P = 0.002). Thus, we conclude that Dby mRNA is selectively retained in capacitated mouse spermatozoa, and it has an important role in the early zygotic development of male mouse zygotes. This might imply that spermatozoa mRNA is involved in early zygotic and embryonic stages of reproduction.
Collapse
|
34
|
|
35
|
Jin XL, O'Neill C. The presence and activation of two essential transcription factors (cAMP response element-binding protein and cAMP-dependent transcription factor ATF1) in the two-cell mouse embryo. Biol Reprod 2009; 82:459-68. [PMID: 19776387 DOI: 10.1095/biolreprod.109.078758] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The expression of two members of an important family of transcription factors, cAMP response element-binding protein (CREB) and cAMP-dependent transcription factor ATF1 (ATF1), is essential for normal preimplantation development. There is a high degree of functional similarity between these two transcription factors, and they can both homodimerize and heterodimerize with each other to form active transcription factors. CREB is present in all stages of mouse preimplantation embryo, and we show here that ATF1 is localized to the nucleus in all preimplantation stages. Activation of these transcription factors requires their phosphorylation, and this was only observed to occur for both transcription factors (serine 133 phosphorylation of CREB and serine 63 phosphorylation of ATF1) at the two-cell stage. Nuclear localization and phosphorylation of ATF1 were constitutive. The nuclear localization and phosphorylation of CREB showed a constitutive component that was further induced by the autocrine embryotropin Paf (1-o-alkyl-2-acetyl-sn-glycero-3-phosphocholine). Activation of CREB by Paf was independent of cAMP but was dependent on calcium, calmodulin, and calmodulin-dependent kinase activity. ATF1 nuclear localization was unaffected by inhibition of the calcium/calmodulin pathway. A complex pattern of expression of calmodulin-dependent kinases was observed throughout preimplantation development. At the two-cell stage, only mRNAs coding for calmodulin-dependent protein kinase kinase beta, calmodulin-dependent protein kinase II gamma, and calmodulin-dependent protein kinase IV were detected. A selective antagonist for calmodulin-dependent protein kinase kinase (STO-609) and calmodulin-dependent protein kinases I, II, and IV (KN-62) blocked the Paf-induced phosphorylation of CREB. The study demonstrates a role for trophic signaling and constitutive activation of two essential transcription factors at the time of zygotic genome activation.
Collapse
Affiliation(s)
- X L Jin
- Human Reproduction Unit, Sydney Centre for Developmental and Regenerative Medicine, Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | | |
Collapse
|
36
|
Wilkerson DC, Sarge KD. RNA polymerase II interacts with the Hspa1b promoter in mouse epididymal spermatozoa. Reproduction 2009; 137:923-9. [PMID: 19336471 DOI: 10.1530/rep-09-0015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The Hspa1b (Hsp70.1) gene is one of the first genes expressed after fertilization, with expression occurring during the minor zygotic genome activation (ZGA) in the absence of stress. This expression can take place in the male pronucleus as early as the one-cell stage of embryogenesis. The importance of HSPA1B for embryonic viability during times of stress is supported by studies showing that depletion of this protein results in a significant reduction in embryos developing to the blastocyte stage. Recently, we have begun addressing the mechanism responsible for allowing expression of Hspa1b during the minor ZGA and found that heat shock transcription factor (HSF) 1 and 2 bind the Hspa1b promoter during late spermatogenesis. In this report, we have extended those studies using western blots and chromatin immunoprecipitation assays and found that RNA polymerase II (Pol II) is present in epididymal spermatozoa and bound to the Hspa1b promoter. These present results, in addition to our previous results, support a model in which the binding of HSF1, HSF2, SP1, and Pol II to the promoter of Hspa1b would allow the rapid formation of a transcription-competent state during the minor ZGA, thereby allowing Hspa1b expression.
Collapse
Affiliation(s)
- Donald C Wilkerson
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536, USA
| | | |
Collapse
|
37
|
SUZUKI T, ABE KI, INOUE A, AOKI F. Expression of c-MYC in Nuclear Speckles During Mouse Oocyte Growth and Preimplantation Development. J Reprod Dev 2009; 55:491-5. [DOI: 10.1262/jrd.09-069a] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Tsukasa SUZUKI
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo
| | - Ken-ichiro ABE
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo
| | - Azusa INOUE
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo
| | - Fugaku AOKI
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo
| |
Collapse
|
38
|
Vigneault C, McGraw S, Sirard MA. Spatiotemporal expression of transcriptional regulators in concert with the maternal-to-embryonic transition during bovine in vitro embryogenesis. Reproduction 2009; 137:13-21. [DOI: 10.1530/rep-08-0077] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cleavage-stage bovine embryos are transcriptionally quiescent until they reach the 8- to 16-cell stage, and thus rely on the reserves provided by the stored maternal mRNAs and proteins found in the oocytes to achieve their first cell divisions. The objective of this study was to characterize the expression and localization of the transcriptional and translational regulators, Y box binding protein 2 (YBX2), TATA box-binding protein (TBP), and activating transcription factor 2 (ATF2), during bovine early embryo development. Germinal vesicle (GV)- and metaphase II (MII)-stage oocytes, as well as 2-, 4-, 8-, 16-cell-stage embryos, morula, and blastocysts, producedin vitrowere analyzed for temporal and spatial protein expression. Using Q-PCR,ATF2mRNA expression was shown to remain constant from the GV-stage oocyte to the four-cell embryo, and then decreased through to the blastocyst stage. By contrast, the protein levels of ATF2 remained constant throughout embryo development and were found in both the cytoplasm and the nucleus. Both TBP and YBX2 showed opposite protein expression patterns, as YBX2 protein levels decreased throughout development, while TBP levels increased through to the blastocyst stage. Immunolocalization studies revealed that TBP protein was localized in the nucleus of 8- to 16-cell-stage embryos, whereas the translational regulator YBX2 was exclusively cytoplasmic and disappeared from the 16-cell stage onward. This study shows that YBX2, TBP, and ATF2 are differentially regulated through embryo development, and provides insight into the molecular events occurring during the activation of the bovine genome during embryo developmentin vitro.
Collapse
|
39
|
Linher K, Cheung Q, Baker P, Bedecarrats G, Shiota K, Li J. An epigenetic mechanism regulates germ cell-specific expression of the porcine Deleted in Azoospermia-Like (DAZL) gene. Differentiation 2008; 77:335-49. [PMID: 19281782 DOI: 10.1016/j.diff.2008.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Revised: 07/10/2008] [Accepted: 08/25/2008] [Indexed: 01/21/2023]
Abstract
The Deleted in Azoospermia-Like (DAZL) gene is specifically expressed in fetal and adult gonads. While DAZL is known to play a role during gametogenesis, the mechanisms governing its germ cell-specific expression remain unclear. We identified the 5' untranslated region (UTR) of the porcine DAZL gene and cloned and characterized 2 kilobase pairs of its TATA-less 5' flanking region, identifying CpG-rich regions within the proximal promoter. Nine of 18 CpG sites in proximity to one region were largely unmethylated in germ cells but hypermethylated in somatic cells, suggesting that DNA methylation may regulate DAZL promoter activity. Furthermore, DAZL expression was induced in fibroblasts treated with a demethylating agent. Deletion analyses revealed that the minimal 149 base pair promoter region was sufficient to activate transcription. In vitro methylation of a reporter construct corresponding to these 149 base pairs resulted in complete suppression of DAZL promoter activity in primordial germ cells, further supporting a role for methylation in regulating DAZL expression. Interestingly, the differentially methylated region was shown to harbor several putative Sp1-binding sites. Mutation of only the most highly conserved site significantly reduced promoter activity in a reporter assay. Furthermore, gel shift assays revealed that Sp1 was able to specifically bind to this site, and that complex formation was inhibited when CpG dinucleotides within this region were methylated. Chromatin immunoprecipitation (ChIP) assays revealed that in vivo Sp1 binding to the core DAZL promoter region was enriched in germ cells but not in fibroblasts. Our data suggests that DNA methylation may suppress DAZL expression in somatic cells by interfering with Sp1 binding. This study provides insights into the potential mechanisms underlying the regulation of germ cell-specific gene expression.
Collapse
Affiliation(s)
- Katja Linher
- Department of Animal and Poultry Science, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | | | | | | | | | | |
Collapse
|
40
|
Activation du génome embryonnaire. ACTA ACUST UNITED AC 2008; 36:1126-32. [DOI: 10.1016/j.gyobfe.2008.07.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Accepted: 07/28/2008] [Indexed: 12/18/2022]
|
41
|
Sun F, Tang F, Yan AY, Fang HY, Sheng HZ. Expression of SRG3, a chromatin-remodelling factor, in the mouse oocyte and early preimplantation embryos. ZYGOTE 2008; 15:129-38. [PMID: 17462105 DOI: 10.1017/s096719940600400x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SRG3 (Smarcc1) is a core subunit of the SWI/SNF complex. In the absence of SRG3, embryonic development ceases during peri-implantation stages, indicating that SRG3, as well as the chromatin-remodelling process, plays an essential role in early mouse development. To gain a better understanding of chromatin remodelling during the early stages of development, we examined SRG3 expression during oogenesis and preimplantation stages using immunofluorescence and western blot assays. SRG3 was detected in nuclei of oocytes during growth and maturation. Following fertilization, SRG3 was detected in pronuclei shortly after their formation. Nuclear concentrations of SRG3 increased in a time-dependent fashion and were found to be greater in the male pronucleus than in the female pronucleus. The increase in nuclear SRG3 was partially inhibited by a protein synthesis inhibitor, but not by a transcriptional inhibitor. Expression of SRG3 is accompanied by expression of Brg1 and Ini1, two other core subunits of the SWI/SNF complex. The expression of these three remodelling factors parallels that of SP1 and TBP, both spatially and temporally, in the mouse embryo, suggesting a role for remodelling factors in chromatin structure and function during early development.
Collapse
Affiliation(s)
- F Sun
- Programme for Graduation Studies, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | | | | | | | | |
Collapse
|
42
|
May A, Reifenberg K, Zechner U, Haaf T. Asynchronous replication dynamics of imprinted and non-imprinted chromosome regions in early mouse embryos. Exp Cell Res 2008; 314:2788-95. [PMID: 18675801 DOI: 10.1016/j.yexcr.2008.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2008] [Revised: 07/13/2008] [Accepted: 07/14/2008] [Indexed: 12/19/2022]
Abstract
We have used interphase FISH to analyze the replication behavior of four imprinted chromosome regions (Snrpn, Zim1-Peg3, Dlk1-Gtl2, and Igf2r) and five non-imprinted regions in mouse one-cell to morula-stage embryos and embryonic fibroblasts. In general, imprinted chromosome regions showed the expected asynchronous pattern of replication throughout all analyzed stages of preimplantation development and in differentiated cells. The Dlk1-Gtl2 locus which is not expressed and Igf2r which is biallelically expressed in early embryos showed a relaxation of replication asynchrony at the morula stage. Asynchronous replication in zygotes and two-cell embryos was not specific to imprinted regions. Three non-imprinted loci (Emp1-Pbp2-Dyntl1, Hbb-b1-Hbb-b2-Hbb-y, and Opa1) as well as one gene-free region on chromosome 7A1 switched from asynchronous replication in one- and two-cell embryos to synchronous replication in 4-cell embryos and later stages. Another gene-free region on chromosome 16C2 showed a more gradual transition from asynchronous to synchronous replication from two-cell to morula-stage embryos. We propose that replication asynchrony contributes to the striking asymmetry between the two parental genomes, which are epigenetically reprogrammed after fertilization into a diploid somatic genome. The switching of non-imprinted genes from asynchronous to synchronous replication may be associated with embryonic genome activation and restoration of transcriptional potential for somatic development.
Collapse
Affiliation(s)
- Andreas May
- Institute for Human Genetics, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | | | | | | |
Collapse
|
43
|
Dadi TD, Li MW, Lloyd KCK. EGF and TGF-alpha supplementation enhances development of cloned mouse embryos. CLONING AND STEM CELLS 2008; 9:315-26. [PMID: 17907942 DOI: 10.1089/clo.2006.0040] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In this study, we sought to determine the extent to which mitogenic growth factors affect the survival and development of cloned mouse embryos in vitro. Cloned embryos derived by intracytoplasmic nuclear injection (ICNI) of cumulus cell nuclei into enucleated oocytes were incubated in culture media supplemented with EGF and/or TGF-alpha for 4 days. Compared to control, treatment with either growth factor significantly increased the blastocyst formation rate, the total number of cells per blastocyst, the cell ratio of the inner cell mass and the trophectoderm (ICM:TE ratio), and EGF-R protein expression in cloned embryos. In most instances these effects were enhanced in cloned embryos when EGF and TGF-alpha were combined. Although fewer blastocysts developed from cloned than from fertilized one-cell stage embryos, growth factor treatment appeared to have the greatest effect on cloned embryos. These results demonstrate that mitogenic growth factors significantly enhance survival and promote the preimplantation development of cloned mouse embryos.
Collapse
Affiliation(s)
- Tedla D Dadi
- Center for Comparative Medicine, School of Veterinary, University of California-Davis, Davis, California 95616, USA.
| | | | | |
Collapse
|
44
|
Ooga M, Inoue A, Kageyama SI, Akiyama T, Nagata M, Aoki F. Changes in H3K79 Methylation During Preimplantation Development in Mice. Biol Reprod 2008; 78:413-24. [DOI: 10.1095/biolreprod.107.063453] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
45
|
Lodde V, Modina S, Maddox-Hyttel P, Franciosi F, Lauria A, Luciano AM. Oocyte morphology and transcriptional silencing in relation to chromatin remodeling during the final phases of bovine oocyte growth. Mol Reprod Dev 2008; 75:915-24. [DOI: 10.1002/mrd.20824] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
46
|
Gazdag E, Rajkovic A, Torres-Padilla ME, Tora L. Analysis of TATA-binding protein 2 (TBP2) and TBP expression suggests different roles for the two proteins in regulation of gene expression during oogenesis and early mouse development. Reproduction 2007; 134:51-62. [PMID: 17641088 DOI: 10.1530/rep-06-0337] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gametogenesis, the process during which germ cells are generated is essential for reproduction. In mammals, maternal mRNA and proteins present in the oocyte are required to ensure the progression of development until the embryo activates its genome after fertilisation. It is well established that the oocyte synthesises these maternal factors during oocyte growth and then undergoes a quiescent transcriptional period that will be resumed only after fertilisation. However, the mechanisms that govern transcriptional regulation and subsequent silencing during oogenesis are not well understood. Here, we have examined the expression and localisation of the TATA-binding protein (TBP) and the related protein TBP2 (also called TRF3, TBP-related factor 3) during oogenesis and in early mouse embryos. We show that TBP is expressed in the oocytes at the beginning of folliculogenesis, but it is undetectable during further stages of oocyte development, and becomes abundant again only after fertilisation. In contrast to TBP, we found that TBP2 is highly expressed in growing oocytes during folliculogenesis, declines upon ovulation, and is almost undetectable after fertilisation by the two-cell stage. The mirroring localisation profile of TBP and TBP2 suggests different roles for the two proteins in establishing specialised programs of gene expression during oocyte development and in early mouse embryos. Analysis of mutant mouse ovaries in which oocyte-specific factors have been knocked-out suggests that TBP2 is a potential candidate for regulating transcriptional control of oogenesis. Moreover, our results obtained with oocytes lacking the oocyte-specific nuclear chaperone nucleoplasmin 2 suggest that TBP2 function may be related to non-condensed chromatin conformation.
Collapse
Affiliation(s)
- Emese Gazdag
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, UMR 7104, CNRS, INSERM, ULP, BP 10142, CU de Strasbourg, 67404 Illkirch Cedex, France
| | | | | | | |
Collapse
|
47
|
Abstract
Mammalian spermatozoa contain some of the most highly compact chromatin. This is due to the DNA binding proteins, the protamines, which replace most of the histones during spermiogenesis. This chromatin, however, shares some features with somatic cell chromatin. One of these is the organization of DNA into loop domains attached at their bases to a proteinaceous nuclear matrix. Several groups have shown that the sites at which DNA associates with the sperm nuclear matrix contain chromatin structures that are linked with specific functions. Recent data also suggest that the sperm nuclear matrix plays essential roles in the paternal pronucleus of the newly fertilized oocyte, suggesting that the sperm cell provides more information to the new embryo than solely the genetic material it delivers. Here, we will review these data which together give insight into the functional significance and requirements of sperm nuclear structure.
Collapse
Affiliation(s)
- Jeffrey A Shaman
- Institute for Biogenesis Research, John A Burns School of Medicine, University of Hawaii. Honolulu, HI 96822, USA
| | | | | |
Collapse
|
48
|
Yamauchi Y, Shaman JA, Boaz SM, Ward WS. Paternal pronuclear DNA degradation is functionally linked to DNA replication in mouse oocytes. Biol Reprod 2007; 77:407-15. [PMID: 17494913 DOI: 10.1095/biolreprod.107.061473] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
We recently demonstrated that mouse spermatozoa contain a mechanism to degrade their DNA into loop-sized fragments of about 50 kb, mediated by topoisomerase IIB, termed sperm chromatin fragmentation (SCF). SCF is often followed by a more complete digestion of the DNA with a sperm nuclease. When SCF-induced spermatozoa are injected into oocytes, the paternal pronuclei degrade their DNA after the initiation of DNA synthesis, but the maternal pronuclei are unaffected and replicate normally. Here, we tested whether the nuclease activity changes in spermatozoa of different maturation stages, and whether there is a functional relationship between the initiation of DNA synthesis and paternal DNA degradation induced by SCF in the zygote. We found that spermatozoa from the vas deferens have a much higher level of SCF activity than those from the cauda epididymis, suggesting that spermatozoa may acquire this activity in the vas deferens. Furthermore, paternal pronuclei formed in zygotes from injecting oocytes with SCF-induced vas deferens spermatozoa degraded their DNA, but this degradation could be inhibited by the DNA synthesis inhibitor, aphidicolin. Upon release from a 4 h aphidicolin-induced arrest, DNA synthesis was initiated in maternal pronuclei, while the paternal pronuclei degraded their DNA. Longer aphidicolin arrest resulted in the paternal pronuclei replicating their DNA, suggesting that delaying the initiation of DNA synthesis allowed the paternal pronuclei to overcome the SCF-induced DNA degradation pathway. These results suggest that the paternal DNA degradation, in oocytes fertilized with SCF-induced spermatozoa, is coupled to the initiation of DNA synthesis in newly fertilized zygotes.
Collapse
Affiliation(s)
- Yasuhiro Yamauchi
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96822, USA
| | | | | | | |
Collapse
|
49
|
Kageyama S, Gunji W, Nakasato M, Murakami Y, Nagata M, Aoki F. Analysis of transcription factor expression during oogenesis and preimplantation development in mice. ZYGOTE 2007; 15:117-28. [PMID: 17462104 DOI: 10.1017/s096719940700411x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SummaryThe transition from a differentiated germ cell into a totipotent zygote during oogenesis and preimplantation development is critical to the creation of a new organism. During this period, cell characteristics change dynamically, suggesting that a global alteration of gene expression patterns occurs, which is regulated by global changes in various epigenetic factors. Among these, transcription factors (TFs) are essential in the direct regulation of transcription and also play important roles in determining cell characteristics. However, no comprehensive analysis of TFs from germ cells to embryos had been undertaken. We used mRNA amplification systems and microarrays to conduct a genomewide analysis of TFs at various stages of oogenesis and preimplantation development. The greatest alteration in TFs occurred between the 1- and 2-cell stages, at which time zygotic genome activation (ZGA) occurs. Our analysis of TFs classified by structure and function revealed several specific patterns of change. Basic transcription factors, which are the general components of transcription, increased transiently at the 2-cell stage, while homeodomain (HD) TFs were expressed specifically in the oocyte. TFs containing the Rel homology region (RHR) and Ets domains were expressed at a high level in 2-cell and blastocyst embryos. Thus, the global TF dynamics that occur during oogenesis and preimplantation development seem to regulate the transition from germ-cell-type to embryo-type gene expression.
Collapse
Affiliation(s)
- S Kageyama
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Chiba 277-8562, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Probst AV, Santos F, Reik W, Almouzni G, Dean W. Structural differences in centromeric heterochromatin are spatially reconciled on fertilisation in the mouse zygote. Chromosoma 2007; 116:403-15. [PMID: 17447080 DOI: 10.1007/s00412-007-0106-8] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 03/07/2007] [Accepted: 03/13/2007] [Indexed: 11/27/2022]
Abstract
In mammals, paternal and maternal pronuclei undergo profound chromatin reorganisation upon fertilisation. How these events are orchestrated within centromeric regions to ensure proper chromosome segregation in the following cellular divisions is unknown. In this study, we followed the dynamic unfolding of the centromeric regions, i.e. the centric and pericentric satellite repeats, by DNA fluorescent in situ hybridization (FISH) during the first cell cycle up to the two-cell stage. The distinct chromatin from female and male gametes both undergo rapid remodelling and reach a zygotic organisation in which the satellites occupy restricted spatial domains surrounding the nucleolar precursor body. A transition from this zygotic to a somatic cell-like organisation takes place during the two-cell stage. Using 3D immuno-FISH, we find that, whereas maternal pericentric regions are marked with H3K9me3, H4K20me3 and HP1beta, paternal ones only showed HP1beta marking. Thus, despite different chromatin features, male and female pronuclei organise their centromeric regions in the same way within the nuclei to align chromosomes on the metaphase plate and segregate them appropriately. Our findings highlight the importance of ensuring a proper centromere function while preserving the distinction of parental genome origin during the return to totipotency in the zygote.
Collapse
Affiliation(s)
- Aline V Probst
- Laboratory of Nuclear Dynamics and Nuclear Plasticity, UMR218 CNRS/Institut Curie, 75248, Paris Cedex 05, France
| | | | | | | | | |
Collapse
|