1
|
Building Pluripotency Identity in the Early Embryo and Derived Stem Cells. Cells 2021; 10:cells10082049. [PMID: 34440818 PMCID: PMC8391114 DOI: 10.3390/cells10082049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 07/27/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
The fusion of two highly differentiated cells, an oocyte with a spermatozoon, gives rise to the zygote, a single totipotent cell, which has the capability to develop into a complete, fully functional organism. Then, as development proceeds, a series of programmed cell divisions occur whereby the arising cells progressively acquire their own cellular and molecular identity, and totipotency narrows until when pluripotency is achieved. The path towards pluripotency involves transcriptome modulation, remodeling of the chromatin epigenetic landscape to which external modulators contribute. Both human and mouse embryos are a source of different types of pluripotent stem cells whose characteristics can be captured and maintained in vitro. The main aim of this review is to address the cellular properties and the molecular signature of the emerging cells during mouse and human early development, highlighting similarities and differences between the two species and between the embryos and their cognate stem cells.
Collapse
|
2
|
Genetics and Epigenetics of One-Carbon Metabolism Pathway in Autism Spectrum Disorder: A Sex-Specific Brain Epigenome? Genes (Basel) 2021; 12:genes12050782. [PMID: 34065323 PMCID: PMC8161134 DOI: 10.3390/genes12050782] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition affecting behavior and communication, presenting with extremely different clinical phenotypes and features. ASD etiology is composite and multifaceted with several causes and risk factors responsible for different individual disease pathophysiological processes and clinical phenotypes. From a genetic and epigenetic side, several candidate genes have been reported as potentially linked to ASD, which can be detected in about 10–25% of patients. Folate gene polymorphisms have been previously associated with other psychiatric and neurodegenerative diseases, mainly focused on gene variants in the DHFR gene (5q14.1; rs70991108, 19bp ins/del), MTHFR gene (1p36.22; rs1801133, C677T and rs1801131, A1298C), and CBS gene (21q22.3; rs876657421, 844ins68). Of note, their roles have been scarcely investigated from a sex/gender viewpoint, though ASD is characterized by a strong sex gap in onset-risk and progression. The aim of the present review is to point out the molecular mechanisms related to intracellular folate recycling affecting in turn remethylation and transsulfuration pathways having potential effects on ASD. Brain epigenome during fetal life necessarily reflects the sex-dependent different imprint of the genome-environment interactions which effects are difficult to decrypt. We here will focus on the DHFR, MTHFR and CBS gene-triad by dissecting their roles in a sex-oriented view, primarily to bring new perspectives in ASD epigenetics.
Collapse
|
3
|
Cai S, Quan S, Yang G, Chen M, Ye Q, Wang G, Yu H, Wang Y, Qiao S, Zeng X. Nutritional Status Impacts Epigenetic Regulation in Early Embryo Development: A Scoping Review. Adv Nutr 2021; 12:1877-1892. [PMID: 33873200 PMCID: PMC8483970 DOI: 10.1093/advances/nmab038] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/03/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
With the increasing maternal age and the use of assisted reproductive technology in various countries worldwide, the influence of epigenetic modification on embryonic development is increasingly notable and prominent. Epigenetic modification disorders caused by various nutritional imbalance would cause embryonic development abnormalities and even have an indelible impact on health in adulthood. In this scoping review, we summarize the main epigenetic modifications in mammals and the synergies among different epigenetic modifications, especially DNA methylation, histone acetylation, and histone methylation. We performed an in-depth analysis of the regulation of various epigenetic modifications on mammals from zygote formation to cleavage stage and blastocyst stage, and reviewed the modifications of key sites and their potential molecular mechanisms. In addition, we discuss the effects of nutrition (protein, lipids, and one-carbon metabolism) on epigenetic modification in embryos and emphasize the importance of various nutrients in embryonic development and epigenetics during pregnancy. Failures in epigenetic regulation have been implicated in mammalian and human early embryo loss and disease. With the use of reproductive technologies, it is becoming even more important to establish developmentally competent embryos. Therefore, it is essential to evaluate the extent to which embryos are sensitive to these epigenetic modifications and nutrition status. Understanding the epigenetic regulation of early embryo development will help us make better use of reproductive technologies and nutrition regulation to improve reproductive health in mammals.
Collapse
Affiliation(s)
- Shuang Cai
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Shuang Quan
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Guangxin Yang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Meixia Chen
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Qianhong Ye
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Gang Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Haitao Yu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Yuming Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | | |
Collapse
|
4
|
Altundag Ö, Çelebi-Saltik B. From Embryo to Adult: One Carbon Metabolism in Stem Cells. Curr Stem Cell Res Ther 2021; 16:175-188. [PMID: 32652922 DOI: 10.2174/1574888x15666200712191308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/16/2020] [Accepted: 04/23/2020] [Indexed: 11/22/2022]
Abstract
Stem cells are undifferentiated cells with self-renewal property and varying differentiation potential that allow the regeneration of tissue cells of an organism throughout adult life beginning from embryonic development. Through the asymmetric cell divisions, each stem cell replicates itself and produces an offspring identical with the mother cell, and a daughter cell that possesses the characteristics of a progenitor cell and commits to a specific lineage to differentiate into tissue cells to maintain homeostasis. To maintain a pool of stem cells to ensure tissue regeneration and homeostasis, it is important to regulate the metabolic functioning of stem cells, progenitor cells and adult tissue stem cells that will meet their internal and external needs. Upon fertilization, the zygote transforms metabolic reprogramming while implantation, embryonic development, organogenesis processes and after birth through adult life. Metabolism in stem cells is a concept that is relatively new to be enlightened. There are no adequate and comprehensive in vitro studies on the comparative analysis of the effects of one-carbon (1-C) metabolism on fetal and adult stem cells compared to embryonic and cancer stem cells' studies that have been reported recently. Since 1-C metabolism is linking parental environmental/ dietary factors and fetal development, investigating the epigenetic, genetic, metabolic and developmental effects on adult period is necessary. Several mutations and abnormalities in 1-C metabolism have been noted in disease changing from diabetes, cancer, pregnancy-related outcomes such as pre-eclampsia, spontaneous abortion, placental abruption, premature delivery, and cardiovascular diseases. In this review, the effects of 1-C metabolism, mainly the methionine and folate metabolism, in stem cells that exist in different developmental stages will be discussed.
Collapse
Affiliation(s)
- Özlem Altundag
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, 06100, Sihhiye, Ankara, Turkey
| | - Betül Çelebi-Saltik
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, 06100, Sihhiye, Ankara, Turkey
| |
Collapse
|
5
|
Zafar MI, Lu S, Li H. Sperm-oocyte interplay: an overview of spermatozoon's role in oocyte activation and current perspectives in diagnosis and fertility treatment. Cell Biosci 2021; 11:4. [PMID: 33407934 PMCID: PMC7789549 DOI: 10.1186/s13578-020-00520-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
The fertilizing spermatozoon is a highly specialized cell that selects from millions along the female tract until the oocyte. The paternal components influence the oocyte activation during fertilization and are fundamental for normal embryo development; however, the sperm-oocyte interplay is in a continuous debate. This review aims to analyze the available scientific information related to the role of the male gamete in the oocyte activation during fertilization, the process of the interaction of sperm factors with oocyte machinery, and the implications of any alterations in this interplay, as well as the advances and limitations of the reproductive techniques and diagnostic tests. At present, both PLCζ and PAWP are the main candidates as oocyte activated factors during fertilization. While PLCζ mechanism is via IP3, how PAWP activates the oocyte still no clear, and these findings are important to study and treat fertilization failure due to oocyte activation, especially when one of the causes is the deficiency of PLCζ in the sperm. However, no diagnostic test has been developed to establish the amount of PLCζ, the protocol to treat this type of pathologies is broad, including treatment with ionophores, sperm selection improvement, and microinjection with PLCζ protein or RNA.
Collapse
Affiliation(s)
- Mohammad Ishraq Zafar
- Institute of Reproductive Health/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, People's Republic of China
| | - Shi Lu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jeifang Avenue, Wuhan, 430022, People's Republic of China
| | - Honggang Li
- Institute of Reproductive Health/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, People's Republic of China. .,Wuhan Tongji Reproductive Medicine Hospital, 128 Sanyang Road, Wuhan, 430013, People's Republic of China.
| |
Collapse
|
6
|
Le Blévec E, Muroňová J, Ray PF, Arnoult C. Paternal epigenetics: Mammalian sperm provide much more than DNA at fertilization. Mol Cell Endocrinol 2020; 518:110964. [PMID: 32738444 DOI: 10.1016/j.mce.2020.110964] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/16/2022]
Abstract
The spermatozoon is a highly differentiated cell with unique characteristics: it is mobile, thanks to its flagellum, and is very compact. The sperm cytoplasm is extremely reduced, containing no ribosomes, and therefore does not allow translation, and its nucleus contains very closed chromatin, preventing transcription. This DNA compaction is linked to the loss of nucleosomes and the replacement of histones by protamines. Based on these characteristics, sperm was considered to simply deliver paternal DNA to the oocyte. However, some parts of the sperm DNA remain organized in a nucleosomal format, and bear epigenetic information. In addition, the nucleus and the cytoplasm contain a multitude of RNAs of different types, including non-coding RNAs (ncRNAs) which also carry epigenetic information. For a long time, these RNAs were considered residues of spermatogenesis. After briefly describing the mechanisms of compaction of sperm DNA, we focus this review on the origin and function of the different ncRNAs. We present studies demonstrating the importance of these RNAs in embryonic development and transgenerational adaptation to stress. We also look at other epigenetic marks, such as DNA methylation or post-translational modifications of histones, and show that they are sensitive to environmental stress and transmissible to offspring. The post-fertilization role of certain sperm-borne proteins is also discussed.
Collapse
Affiliation(s)
- Emilie Le Blévec
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France; IMV Technologies, ZI N° 1 Est, L'Aigle, F-61300, France
| | - Jana Muroňová
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France
| | - Pierre F Ray
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France; CHU de Grenoble, UM GI-DPI, Grenoble, F-38000, France
| | - Christophe Arnoult
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France.
| |
Collapse
|
7
|
Raad G, Azouri J, Rizk K, Zeidan NS, Azouri J, Grandjean V, Hazzouri M. Adverse effects of paternal obesity on the motile spermatozoa quality. PLoS One 2019; 14:e0211837. [PMID: 30742661 PMCID: PMC6370200 DOI: 10.1371/journal.pone.0211837] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022] Open
Abstract
Growing evidence suggests that paternal obesity may decrease male fertility potential. During infertility treatment with intra-cytoplasmic sperm injection (ICSI), a morphologically normal motile spermatozoon is injected into a mature egg, when possible. However, sperm motility and morphology per se do not reflect the sperm molecular composition. In this study, we aimed to assess the quality of motile spermatozoa in the context of obesity by analysing their conventional and molecular characteristics as well as their ability to promote early embryonic development. A prospective study was conducted on 128 infertile men divided into three groups: 40 lean, 42 overweight, and 46 obese men. Conventional sperm parameters (concentration, motility and morphology) and sperm molecular status (chromatin composition and integrity, 5-methycytosine (5-mC) and 5-hydroxycytosine (5-hmC) contents and oxidative stress level) were analysed on raw semen and/or on motile spermatozoa selected by density gradient or swim-up techniques. Morphokinetic analysis of the embryos derived from ICSI was performed using the Embryoviewer software. Our results showed that the motile sperm-enriched fraction from obese men exhibited higher levels of retained histones (p<0.001), elevated percentage of altered chromatin integrity (p<0.001), and decreased contents of 5-hmC (p<0.001), and 5-mC (p<0.05) levels as compared to that from lean men. Importantly, there were no statistically significant correlations between these molecular parameters and the percentages of morphologically normal motile spermatozoa. Regarding embryo morphokinetics, the CC1 (p<0.05) and CC3 (p<0.05) embryonic cell cycles were significantly delayed in the cleavage embryos of the obese group as compared to the embryos of the lean group. Our data is of particular interest because, besides demonstrating the negative impacts of obesity on motile spermatozoa molecular composition, it also highlights the possible risk of disturbing early embryonic cell cycles kinetics in the context of paternal obesity.
Collapse
Affiliation(s)
- Georges Raad
- Azoury-IVF clinic, Mount Lebanon Hospital, Camille Chamoun Boulevard, Beirut, Lebanon
- Lebanese University, Faculty of Sciences 2, Fanar, Lebanon
| | - Joseph Azouri
- Azoury-IVF clinic, Mount Lebanon Hospital, Camille Chamoun Boulevard, Beirut, Lebanon
| | - Kamal Rizk
- Azoury-IVF clinic, Mount Lebanon Hospital, Camille Chamoun Boulevard, Beirut, Lebanon
| | - Nina S. Zeidan
- Lebanese University, Faculty of Sciences 2, Fanar, Lebanon
| | - Jessica Azouri
- Azoury-IVF clinic, Mount Lebanon Hospital, Camille Chamoun Boulevard, Beirut, Lebanon
| | - Valérie Grandjean
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 10 “Control of gene expression “, Nice, France and University of Nice Sophia Antipolis, Faculty of Medecine, Nice, France
- * E-mail:
| | - Mira Hazzouri
- Lebanese University, Faculty of Sciences 2, Fanar, Lebanon
| |
Collapse
|
8
|
Early post-conception maternal cortisol, children’s HPAA activity and DNA methylation profiles. J Dev Orig Health Dis 2018; 10:73-87. [DOI: 10.1017/s2040174418000880] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AbstractThe hypothalamic–pituitary–adrenal axis (HPAA) plays a critical role in the functioning of all other biological systems. Thus, studying how the environment may influence its ontogeny is paramount to understanding developmental origins of health and disease. The early post-conceptional (EPC) period could be particularly important for the HPAA as the effects of exposures on organisms’ first cells can be transmitted through all cell lineages. We evaluate putative relationships between EPC maternal cortisol levels, a marker of physiologic stress, and their children’s pre-pubertal HPAA activity (n=22 dyads). Maternal first-morning urinary (FMU) cortisol, collected every-other-day during the first 8 weeks post-conception, was associated with children’s FMU cortisol collected daily around the start of the school year, a non-experimental challenge, as well as salivary cortisol responses to an experimental challenge (all Ps<0.05), with some sex-related differences. We investigated whether epigenetic mechanisms statistically mediated these links and, therefore, could provide cues as to possible biological pathways involved. EPC cortisol was associated with >5% change in children’s buccal epithelial cells’ DNA methylation for 867 sites, while children’s HPAA activity was associated with five CpG sites. Yet, no CpG sites were related to both, EPC cortisol and children’s HPAA activity. Thus, these epigenetic modifications did not statistically mediate the observed physiological links. Larger, prospective peri-conceptional cohort studies including frequent bio-specimen collection from mothers and children will be required to replicate our analyses and, if our results are confirmed, identify biological mechanisms mediating the statistical links observed between maternal EPC cortisol and children’s HPAA activity.
Collapse
|
9
|
Suzuki S, Minami N. CHD1 Controls Cell Lineage Specification Through Zygotic Genome Activation. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2018; 229:15-30. [PMID: 29177762 DOI: 10.1007/978-3-319-63187-5_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In mammals, the processes spanning from fertilization to the generation of a new organism are very complex and are controlled by multiple genes. Life begins with the encounter of eggs and spermatozoa, in which gene expression is inactive prior to fertilization. After several cell divisions, cells arise that are specialized in implantation, a developmental process unique to mammals. Cells involved in the establishment and maintenance of implantation differentiate from totipotent embryos, and the remaining cells generate the embryo proper. Although this process of differentiation, termed cell lineage specification, is supported by various gene expression networks, many components have yet to be identified. Moreover, despite extensive research it remains unclear which genes are controlled by each of the factors involved. Although it has become clear that epigenetic factors regulate gene expression, elucidation of the underlying mechanisms remains challenging. In this chapter, we propose that the chromatin remodeling factor CHD1, together with epigenetic factors, is involved in a subset of gene expression networks involved in processes spanning from zygotic genome activation to cell lineage specification.
Collapse
Affiliation(s)
- Shinnosuke Suzuki
- Technology and Development Team for Mammalian Genome Dynamics, RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Naojiro Minami
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8052, Japan.
| |
Collapse
|
10
|
Xu J, Sinclair KD. One-carbon metabolism and epigenetic regulation of embryo development. Reprod Fertil Dev 2017; 27:667-76. [PMID: 25710200 DOI: 10.1071/rd14377] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/08/2015] [Indexed: 12/15/2022] Open
Abstract
One-carbon (1C) metabolism consists of an integrated series of metabolic pathways that include the folate cycle and methionine remethylation and trans-sulfuration pathways. Most, but not all, 1C metabolic enzymes are expressed in somatic cells of the ovary, mammalian oocytes and in preimplantation embryos. The metabolic implications of this, with regard to the provision of methyl donors (e.g. betaine) and 1C cofactors (e.g. vitamin B12), together with consequences of polymorphic variances in genes encoding 1C enzymes, are not fully understood but are the subject of ongoing investigations at the authors' laboratory. However, deficiencies in 1C-related substrates and/or cofactors during the periconception period are known to lead to epigenetic alterations in DNA and histone methylation in genes that regulate key developmental processes in the embryo. Such epigenetic modifications have been demonstrated to negatively impact on the subsequent health and metabolism of offspring. For this reason, parental nutrition around the time of conception has become a focal point of investigation in many laboratories with the aim of providing improved nutritional advice to couples. These issues are considered in detail in this article, which offers a contemporary overview of the effects of 1C metabolism on epigenetic programming in mammalian gametes and the early embryo.
Collapse
Affiliation(s)
- Juan Xu
- School of Bioscience, University of Nottingham, Sutton Bonington, Leicestershire LE12 5RD, UK
| | - Kevin D Sinclair
- School of Bioscience, University of Nottingham, Sutton Bonington, Leicestershire LE12 5RD, UK
| |
Collapse
|
11
|
Abstract
IMPORTANCE In recent decades, a trend toward delayed childbearing is noted in developed countries. Whereas the effects of maternal age on fertility, pregnancy complications, and postnatal outcomes have been thoroughly explored, consequences of advanced paternal age are less well known. Oocyte donation cycles can be used as an optimal model to analyze the association between male ageing and reproductive outcomes with minimal confounding. OBJECTIVE The purpose of this work was to summarize the updated and relevant literature dealing with the effect of paternal age on oocyte donation outcomes. RESULTS According to the available evidence from oocyte donation cycles, it seems that no significant association exists between advanced paternal age and fertility. However, this evidence is based on few studies, many of which are of low quality, yielding conflicting results. In addition, the emerging evidence clearly indicates an increased risk of adverse postnatal manifestations of pregnancies conceived by older fathers, including de novo autosomal dominant disorders, impaired neurocognitive development, and increased risk of malignancy. CONCLUSIONS AND RELEVANCE This review may be of aid to medical practitioners in counseling couples on the risks of delayed childbearing.
Collapse
|
12
|
Baroux C, Grossniklaus U. The Maternal-to-Zygotic Transition in Flowering Plants: Evidence, Mechanisms, and Plasticity. Curr Top Dev Biol 2015; 113:351-71. [PMID: 26358878 DOI: 10.1016/bs.ctdb.2015.06.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The maternal-to-zygotic transition (MZT) defines a developmental phase during which the embryo progressively emancipates itself from a developmental control relying largely on maternal information. The MZT is a functional readout of two processes: the clearance of maternally derived information and the de novo expression of the inherited, parental alleles enabled by zygotic genome activation (ZGA). In plants, for many years the debate about whether the MZT exists at all focused on the ZGA alone. However, several recent studies provide evidence for a progressive alleviation of the maternal control over embryogenesis that is correlated with a gradual ZGA, a process that is itself maternally controlled. Yet, several examples of zygotic genes that are expressed and/or functionally required early in embryogenesis demonstrate a certain flexibility in the dynamics and kinetics of the MZT among plant species and also intraspecific hybrids.
Collapse
Affiliation(s)
- Célia Baroux
- Institute of Plant Biology & Zurich-Basel Plant Science Center, University of Zurich, Zurich, Switzerland
| | - Ueli Grossniklaus
- Institute of Plant Biology & Zurich-Basel Plant Science Center, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
Suzuki S, Nozawa Y, Tsukamoto S, Kaneko T, Manabe I, Imai H, Minami N. CHD1 acts via the Hmgpi pathway to regulate mouse early embryogenesis. Development 2015; 142:2375-84. [PMID: 26092847 DOI: 10.1242/dev.120493] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 05/14/2015] [Indexed: 11/20/2022]
Abstract
The protein CHD1 is a member of the family of ATPase-dependent chromatin remodeling factors. CHD1, which recognizes trimethylated histone H3 lysine 4, has been implicated in transcriptional activation in organisms ranging from yeast to humans. It is required for pre-mRNA maturation, maintenance of mouse embryonic stem cell pluripotency and rapid growth of the mouse epiblast. However, the function(s) of CHD1 in mouse preimplantation embryos has not yet been examined. Here, we show that loss of CHD1 function led to embryonic lethality after implantation. In mouse embryos in which Chd1 was targeted by siRNA microinjection, the expression of the key regulators of cell fate specification Pou5f1 (also known as Oct4), Nanog and Cdx2 was dramatically decreased, starting at mid-preimplantation gene activation (MGA). Moreover, expression of Hmgpi and Klf5, which regulate Pou5f1, Nanog and Cdx2, was also significantly suppressed at zygotic gene activation (ZGA). Suppression of Hmgpi expression in Chd1-knockdown embryos continued until the blastocyst stage, whereas suppression of Klf5 expression was relieved by the morula stage. Next, we rescued HMGPI expression via Hmgpi mRNA microinjection in Chd1-knockdown embryos. Consequently, Pou5f1, Nanog and Cdx2 expression was restored at MGA and live offspring were recovered. These findings indicate that CHD1 plays important roles in mouse early embryogenesis via activation of Hmgpi at ZGA.
Collapse
Affiliation(s)
- Shinnosuke Suzuki
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Yusuke Nozawa
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Satoshi Tsukamoto
- Laboratory of Animal and Genome Sciences Section, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Takehito Kaneko
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Ichiro Manabe
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroshi Imai
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Naojiro Minami
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
14
|
Beaujean N. Epigenetics, embryo quality and developmental potential. Reprod Fertil Dev 2015; 27:53-62. [DOI: 10.1071/rd14309] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
It is very important for embryologists to understand how parental inherited genomes are reprogrammed after fertilisation in order to obtain good-quality embryos that will sustain further development. In mammals, it is now well established that important epigenetic modifications occur after fertilisation. Although gametes carry special epigenetic signatures, they should attain embryo-specific signatures, some of which are crucial for the production of healthy embryos. Indeed, it appears that proper establishment of different epigenetic modifications and subsequent scaffolding of the chromatin are crucial steps during the first cleavages. This ‘reprogramming’ is promoted by the intimate contact between the parental inherited genomes and the oocyte cytoplasm after fusion of the gametes. This review introduces two main epigenetic players, namely histone post-translational modifications and DNA methylation, and highlights their importance during early embryonic development.
Collapse
|
15
|
Salvaing J, Li Y, Beaujean N, O'Neill C. Determinants of valid measurements of global changes in 5ʹ-methylcytosine and 5ʹ-hydroxymethylcytosine by immunolocalisation in the early embryo. Reprod Fertil Dev 2015; 27:755-64. [DOI: 10.1071/rd14136] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 08/26/2014] [Indexed: 12/15/2022] Open
Abstract
A classical model of epigenetic reprogramming of methyl-cytosine–phosphate–guanine (CpG) dinucleotides within the genome of the early embryo involves a process of active demethylation of the paternally derived genome immediately following fertilisation, creating marked asymmetry in global cytosine methylation levels in male and female pronuclei, followed by passive demethylation of the maternally derived genome over subsequent cell cycles. This model has dominated thinking in developmental epigenetics over recent decades. Recent re-analyses of the model show that demethylation of the paternally derived genome is more modest than formerly thought and results in overall similar levels of methylation of the paternal and maternal pronuclei in presyngamal zygotes, although there is little evidence for a pervasive process of passive demethylation during the cleavage stage of development. In contrast, the inner cell mass of the blastocyst shows some loss of methylation within specific classes of loci. Improved methods of chemical analysis now allow global base-level analysis of modifications to CpG dinucleotides within the cells of the early embryo, yet the low cost and convenience of the immunolocalisation techniques mean that they still have a valuable place in the analysis of the epigenetics of embryo development. In this review we consider the key strengths and weaknesses of this methodology and some factors required for its valid use and interpretation.
Collapse
|
16
|
Efimova OA, Pendina AA, Tikhonov AV, Fedorova ID, Krapivin MI, Chiryaeva OG, Shilnikova EM, Bogdanova MA, Kogan IY, Kuznetzova TV, Gzgzyan AM, Ailamazyan EK, Baranov VS. Chromosome hydroxymethylation patterns in human zygotes and cleavage-stage embryos. Reproduction 2014; 149:223-33. [PMID: 25504867 DOI: 10.1530/rep-14-0343] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We report the sequential changes in 5-hydroxymethylcytosine (5hmC) patterns in the genome of human preimplantation embryos during DNA methylation reprogramming. We have studied chromosome hydroxymethylation and methylation patterns in triploid zygotes and blastomeres of cleavage-stage embryos. Using indirect immunofluorescence, we have analyzed the localization of 5hmC and its co-distribution with 5-methylcytosine (5mC) on the QFH-banded metaphase chromosomes. In zygotes, 5hmC accumulates in both parental chromosome sets, but hydroxymethylation is more intensive in the poorly methylated paternal set. In the maternal set, chromosomes are highly methylated, but contain little 5hmC. Hydroxymethylation is highly region specific in both parental chromosome sets: hydroxymethylated loci correspond to R-bands, but not G-bands, and have well-defined borders, which coincide with the R/G-band boundaries. The centromeric regions and heterochromatin at 1q12, 9q12, 16q11.2, and Yq12 contain little 5mC and no 5hmC. We hypothesize that 5hmC may mark structural/functional genome 'units' corresponding to chromosome bands in the newly formed zygotic genome. In addition, we suggest that the hydroxymethylation of R-bands in zygotes can be treated as a new characteristic distinguishing them from G-bands. At cleavages, chromosomes with asymmetrical hydroxymethylation of sister chromatids appear. They decrease in number during cleavages, whereas totally non-hydroxymethylated chromosomes become numerous. Taken together, our findings suggest that, in the zygotic genome, 5hmC is distributed selectively and its pattern is determined by both parental origin of chromosomes and type of chromosome bands - R, G, or C. At cleavages, chromosome hydroxymethylation pattern is dynamically changed due to passive and non-selective overall loss of 5hmC, which coincides with that of 5mC.
Collapse
Affiliation(s)
- Olga A Efimova
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia
| | - Anna A Pendina
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Pet
| | - Andrei V Tikhonov
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Pet
| | - Irina D Fedorova
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia
| | - Mikhail I Krapivin
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia
| | - Olga G Chiryaeva
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Pet
| | - Evgeniia M Shilnikova
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia
| | - Mariia A Bogdanova
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia
| | - Igor Yu Kogan
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia
| | - Tatyana V Kuznetzova
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia
| | - Alexander M Gzgzyan
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia
| | - Edward K Ailamazyan
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Pet
| | - Vladislav S Baranov
- D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Petersburg, Russia D.O. Ott Research Institute of Obstetrics and GynecologyMendeleevskaya line, 3, 199034 St Petersburg, RussiaSt Petersburg State UniversityUniversitetskaya nab.7/9, 199034 St Petersburg, RussiaCenter for Medical GeneticsTobolskaya ul., 5, 194044 St Petersburg, RussiaSt Petersburg State Pediatric Medical UniversityLitovskaya ul., 2, 194100 St Petersburg, RussiaS.M. Kirov Military Medical AcademyLebedeva ul., 6, 194044 St Petersburg, RussiaN.I. Pirogov National Medical-Surgery CenterSt Petersburg Clinic Complex, nab. Fontanki, 154, 190103 St Petersburg, RussiaI.P. Pavlov First St Petersburg State Medical UniversityL'va Tolstogo ul., 6/8, 197022 St Pet
| |
Collapse
|
17
|
Guz J, Gackowski D, Foksinski M, Rozalski R, Olinski R. Comparison of the Absolute Level of Epigenetic Marks 5-Methylcytosine, 5-Hydroxymethylcytosine, and 5-Hydroxymethyluracil Between Human Leukocytes and Sperm1. Biol Reprod 2014; 91:55. [DOI: 10.1095/biolreprod.114.121541] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
18
|
Messerschmidt DM, Knowles BB, Solter D. DNA methylation dynamics during epigenetic reprogramming in the germline and preimplantation embryos. Genes Dev 2014; 28:812-28. [PMID: 24736841 PMCID: PMC4003274 DOI: 10.1101/gad.234294.113] [Citation(s) in RCA: 463] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Methylation of DNA is an essential epigenetic control mechanism in mammals. Messerschmidt et al. review the current understanding of epigenetic dynamics regulating the molecular processes that prepare the mammalian embryo for normal development. Methylation of DNA is an essential epigenetic control mechanism in mammals. During embryonic development, cells are directed toward their future lineages, and DNA methylation poses a fundamental epigenetic barrier that guides and restricts differentiation and prevents regression into an undifferentiated state. DNA methylation also plays an important role in sex chromosome dosage compensation, the repression of retrotransposons that threaten genome integrity, the maintenance of genome stability, and the coordinated expression of imprinted genes. However, DNA methylation marks must be globally removed to allow for sexual reproduction and the adoption of the specialized, hypomethylated epigenome of the primordial germ cell and the preimplantation embryo. Recent technological advances in genome-wide DNA methylation analysis and the functional description of novel enzymatic DNA demethylation pathways have provided significant insights into the molecular processes that prepare the mammalian embryo for normal development.
Collapse
Affiliation(s)
- Daniel M Messerschmidt
- Developmental Epigenetics and Disease, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), 138673 Singapore,
| | | | | |
Collapse
|
19
|
Li L, Lu X, Dean J. The maternal to zygotic transition in mammals. Mol Aspects Med 2013; 34:919-38. [PMID: 23352575 DOI: 10.1016/j.mam.2013.01.003] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 01/08/2013] [Accepted: 01/11/2013] [Indexed: 11/15/2022]
Abstract
Prior to activation of the embryonic genome, the initiating events of mammalian development are under maternal control and include fertilization, the block to polyspermy and processing sperm DNA. Following gamete union, the transcriptionally inert sperm DNA is repackaged into the male pronucleus which fuses with the female pronucleus to form a 1-cell zygote. Embryonic transcription begins during the maternal to zygotic transfer of control in directing development. This transition occurs at species-specific times after one or several rounds of blastomere cleavage and is essential for normal development. However, even after activation of the embryonic genome, successful development relies on stored maternal components without which embryos fail to progress beyond initial cell divisions. Better understanding of the molecular basis of maternal to zygotic transition including fertilization, the activation of the embryonic genome and cleavage-stage development will provide insight into early human development that should translate into clinical applications for regenerative medicine and assisted reproductive technologies.
Collapse
Affiliation(s)
- Lei Li
- Division of Molecular Embryonic Development, State Key Laboratory of Reproductive Biology, Institute of Zoology/Chinese Academy of Sciences, Beijing 100101, PR China.
| | | | | |
Collapse
|
20
|
Li F, Li L, Zhong Y, Xie Q, Huang J, Kang X, Wang D, Xu L, Huang T. Relationship between LTR methylation and gag expression of HIV-1 in human spermatozoa and sperm-derived embryos. PLoS One 2013; 8:e54801. [PMID: 23382972 PMCID: PMC3557281 DOI: 10.1371/journal.pone.0054801] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 12/17/2012] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Studying the methylation status of long terminal repeats (LTR) and its relationship to gag expression of HIV-1 in order to explore regulation mechanism of HIV-1 gene expression in vertical transmission from sperm to embryo. METHODS/PRINCIPAL FINDINGS Sperm samples were collected from a healthy donor and seven patients with HIV/AIDS. Zona-free hamster ova were fertilized by donor's spermatozoa transfected with pIRES2-EGFP-LTR-gag and patient's spermatozoa to obtain zygotes and 2-cell embryos, respectively. Interspecific in vitro fertilization, bisulfite sequencing PCR (BSP), RT-PCR, nested RT-PCR, nested real-time qRT-PCR and 2(-△△Ct) method, indirect immunofluoresence (IF) assay were performed. For donor's samples, the methylation rates of HIV-1 LTR were 0.56%, 1.67%, 0.56%, 0.56% in plasmid, spermatozoa, zygotes and 2-cell embryos, respectively while spermatozoa were transfected with unmethylated plasmid, and were 95.0%, 84.44%, 3.3%, 1.67% while transfected with methylated plasmid. The positive bands for HIV-1 gag cDNA were detected in spermatozoa and 2-cell embryos. The positive signals for HIV-1 p24 Gag protein were detected in 2-cell embryos but not in spermatozoa. For patient's samples, methylation rates of HIV-1 LTR were different in spermatozoa among patients. After fertilization, CpG sites in HIV-1 LTR were highly demethylated in zygotes and 2-cell embryos. The gag transcription levels increased with decreasing of methylation rates of HIV-1 LTR, which showed a strong negative correlations between gag transcription levels and methylation rates of HIV-LTR ether in the spermatozoa (r = -0.9877, P<0.0001) or in the sperm-derived 2-cell embryos (r = -0.9092, P = 0.0045). CONCLUSION LTR methylation regulates expression of HIV-1 gag in vertical transmission from sperm to embryo.
Collapse
Affiliation(s)
- FangZheng Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Research Center for Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - LianBing Li
- Key Laboratory of Birth Defects and Reproductive Health, Chongqing, China
| | - Ying Zhong
- Center for Reproductive Medicine, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu, Sichuan, China
| | - QingDong Xie
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Research Center for Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - JiHua Huang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Research Center for Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - XiangJin Kang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Research Center for Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Dian Wang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Research Center for Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Lan Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Research Center for Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - TianHua Huang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Research Center for Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong, China
- * E-mail:
| |
Collapse
|
21
|
Abstract
Since the early twentieth century, inheritance was seen as the inheritance of genes. Concurrent with the acceptance of the genetic theory of inheritance was the rejection of the idea that the cytoplasm of the oocyte could also play a role in inheritance and a corresponding devaluation of embryology as a discipline critical for understanding human development. Development, and variation in development, came to be viewed solely as matters of genetic inheritance and genetic variation. We now know that inheritance is a matter of both genetic and cytoplasmic inheritance. A growing awareness of the centrality of the cytoplasm in explaining both human development and phenotypic variation has been promoted by two contemporaneous developments: the continuing elaboration of the molecular mechanisms of epigenetics and the global rise of artificial reproductive technologies. I review recent developments in the ongoing elaboration of the role of the cytoplasm in human inheritance and development.
Collapse
Affiliation(s)
- Evan Charney
- Sanford School of Public Policy, Duke University, Durham, NC, USA.
| |
Collapse
|
22
|
Zeybel M, Hardy T, Wong YK, Mathers JC, Fox CR, Gackowska A, Oakley F, Burt AD, Wilson CL, Anstee QM, Barter MJ, Masson S, Elsharkawy AM, Mann DA, Mann J. Multigenerational epigenetic adaptation of the hepatic wound-healing response. Nat Med 2012; 18:1369-77. [PMID: 22941276 PMCID: PMC3489975 DOI: 10.1038/nm.2893] [Citation(s) in RCA: 203] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 07/02/2012] [Indexed: 02/06/2023]
Abstract
We investigated whether ancestral liver damage leads to heritable reprogramming of hepatic wound healing in male rats. We found that a history of liver damage corresponds with transmission of an epigenetic suppressive adaptation of the fibrogenic component of wound healing to the male F1 and F2 generations. Underlying this adaptation was less generation of liver myofibroblasts, higher hepatic expression of the antifibrogenic factor peroxisome proliferator-activated receptor γ (PPAR-γ) and lower expression of the profibrogenic factor transforming growth factor β1 (TGF-β1) compared to rats without this adaptation. Remodeling of DNA methylation and histone acetylation underpinned these alterations in gene expression. Sperm from rats with liver fibrosis were enriched for the histone variant H2A.Z and trimethylation of histone H3 at Lys27 (H3K27me3) at PPAR-γ chromatin. These modifications to the sperm chromatin were transmittable by adaptive serum transfer from fibrotic rats to naive rats and similar modifications were induced in mesenchymal stem cells exposed to conditioned media from cultured rat or human myofibroblasts. Thus, it is probable that a myofibroblast-secreted soluble factor stimulates heritable epigenetic signatures in sperm so that the resulting offspring better adapt to future fibrogenic hepatic insults. Adding possible relevance to humans, we found that people with mild liver fibrosis have hypomethylation of the PPARG promoter compared to others with severe fibrosis.
Collapse
Affiliation(s)
- Müjdat Zeybel
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Jenkins TG, Carrell DT. Dynamic alterations in the paternal epigenetic landscape following fertilization. Front Genet 2012; 3:143. [PMID: 23024648 PMCID: PMC3442791 DOI: 10.3389/fgene.2012.00143] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/13/2012] [Indexed: 12/14/2022] Open
Abstract
Embryonic development is a complex and dynamic process with frequent changes in gene expression, ultimately leading to cellular differentiation and commitment of various cell lines. These changes are likely preceded by changes to signaling cascades and/or alterations to the epigenetic program in specific cells. The process of epigenetic remodeling begins early in development. In fact, soon after the union of sperm and egg massive epigenetic changes occur across the paternal and maternal epigenetic landscape. The epigenome of these cells includes modifications to the DNA itself, in the form of DNA methylation, as well as nuclear protein content and modification, such as modifications to histones. Sperm chromatin is predominantly packaged by protamines, but following fertilization the sperm pronucleus undergoes remodeling in which maternally derived histones replace protamines, resulting in the relaxation of chromatin and ultimately decondensation of the paternal pronucleus. In addition, active DNA demethylation occurs across the paternal genome prior to the first cell division, effectively erasing many spermatogenesis derived methylation marks. This complex interplay begins the dynamic process by which two haploid cells unite to form a diploid organism. The biology of these events is central to the understanding of sexual reproduction, yet our knowledge regarding the mechanisms involved is extremely limited. This review will explore what is known regarding the post-fertilization epigenetic alterations of the paternal chromatin and the implications suggested by the available literature.
Collapse
Affiliation(s)
- Timothy G Jenkins
- Andrology and IVF Laboratories, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | |
Collapse
|
24
|
Abstract
Understanding the molecular mechanisms involved in the control of cell differentiation during embryonic development is currently one of the main objectives of developmental biology. This knowledge will provide a basis for the development of new strategies in the field of regenerative medicine, one of the most promising weapons to fight many human diseases. Cell differentiation during embryonic development is controlled primarily by epigenetic factors, that is, mechanisms involved in the regulation of chromatin structure and gene expression. Here we describe the best known epigenetic modifications, and pathways, mainly focused on DNA methylation and histone modifications, and try to depict the state of art in our knowledge about epigenetic regulation of embryonic stem cell maintenance and differentiation.
Collapse
|
25
|
Calvanese V, Lara E, Fraga MF. Epigenetic code and self-identity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 738:236-55. [PMID: 22399383 DOI: 10.1007/978-1-4614-1680-7_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Epigenetics is a new and expanding science that studies the chromatin-based regulation of gene expression. It is achieving considerable importance, especially with regard to developmental mechanisms that drive cell and organ differentiation, as well as in all those biological processes that involve response and adaptation to environmental stimuli. One of the most interesting biological questions concerning animals, especially human beings, is the ability to distinguish self from nonself. This ability has developed throughout evolution, both as the main function of the immune system, which defends against attack by foreign organisms and at the level of consciousness of oneself as an individual, one of the highest functions of the brain that enables social life. Here we will attempt to dissect the epigenetic mechanisms involved in establishing these higher functions and describe some alterations of the epigenetic machinery responsible for the impairment of correct self-recognition and self-identity.
Collapse
Affiliation(s)
- Vincenzo Calvanese
- Department of Immunology and Oncology, National Center for Biotechnology, Madrid, Spain
| | | | | |
Collapse
|
26
|
Zhang RP, Shao JZ, Xiang LX. GADD45A protein plays an essential role in active DNA demethylation during terminal osteogenic differentiation of adipose-derived mesenchymal stem cells. J Biol Chem 2011; 286:41083-94. [PMID: 21917922 PMCID: PMC3220515 DOI: 10.1074/jbc.m111.258715] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Revised: 09/14/2011] [Indexed: 12/13/2022] Open
Abstract
Methylation and demethylation of DNA are the complementary processes of epigenetic regulation. These two types of regulation influence a diverse array of cellular activities, including the maintenance of pluripotency and self-renewal in embryonic stem cells. It was generally believed that DNA demethylation occurs passively over several cycles of DNA replication and that active DNA demethylation is rare. Recently, evidence for active DNA demethylation has been obtained in several cancer, neuronal, and embryonic stem cell lines. Studies in embryonic stem cell models, however, suggested that active DNA demethylation might be restricted to the early development of progenitor cells. Whether active demethylation is involved in terminal differentiation of adult stem cells is poorly understood. We provide evidence that active DNA demethylation does occur during terminal specification of stem cells in an adipose-derived mesenchymal stem cell-derived osteogenic differentiation model. The medium CpG regions in promoters of the Dlx5, Runx2, Bglap, and Osterix osteogenic lineage-specific genes were demethylated during the increase in gene expression associated with osteogenic differentiation. The growth arrest and DNA damage-inducible protein GADD45A was up-regulated in these processes. Knockdown of GADD45A led to hypermethylation of Dlx5, Runx2, Bglap, and Osterix promoters, followed by suppression of the expression of these genes and interruption of osteogenic differentiation. These results reveal that GADD45A plays an essential role in gene-specific active DNA demethylation during adult stem cell differentiation. They enhance the current knowledge of osteogenic specification and may also lead to a better understanding of the common mechanisms underlying epigenetic regulation in adult stem cell differentiation.
Collapse
Affiliation(s)
- Rui-peng Zhang
- From the College of Life Sciences, Zhejiang University, Hangzhou 310058 and
- the Key Laboratory for Cell and Gene Engineering, Hangzhou 310058, Zhejiang Province, China
| | - Jian-zhong Shao
- From the College of Life Sciences, Zhejiang University, Hangzhou 310058 and
- the Key Laboratory for Cell and Gene Engineering, Hangzhou 310058, Zhejiang Province, China
| | - Li-xin Xiang
- From the College of Life Sciences, Zhejiang University, Hangzhou 310058 and
- the Key Laboratory for Cell and Gene Engineering, Hangzhou 310058, Zhejiang Province, China
| |
Collapse
|
27
|
Milosavljevic A. Emerging patterns of epigenomic variation. Trends Genet 2011; 27:242-50. [PMID: 21507501 DOI: 10.1016/j.tig.2011.03.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 03/09/2011] [Accepted: 03/14/2011] [Indexed: 12/15/2022]
Abstract
Fuelled by new sequencing technologies, epigenome mapping projects are revealing epigenomic variation at all levels of biological complexity, from species to cells. Comparisons of methylation profiles among species reveal evolutionary conservation of gene body methylation patterns, pointing to the fundamental role of epigenomes in gene regulation. At the human population level, epigenomic changes provide footprints of the effects of genomic variants within the vast nonprotein-coding fraction of the genome, and comparisons of the epigenomes of parents and their offspring point to quantitative epigenomic parent-of-origin effects confounding classical Mendelian genetics. At the organismal level, comparisons of epigenomes from diverse cell types provide insights into cellular differentiation. Finally, comparisons of epigenomes from monozygotic twins help dissect genetic and environmental influences on human phenotypes and longitudinal comparisons reveal aging-associated epigenomic drift. The development of new bioinformatic frameworks for comparative epigenome analysis is putting epigenome maps within the reach of researchers across a wide spectrum of biological disciplines.
Collapse
|
28
|
Hales BF, Grenier L, Lalancette C, Robaire B. Epigenetic programming: From gametes to blastocyst. ACTA ACUST UNITED AC 2011; 91:652-65. [DOI: 10.1002/bdra.20781] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 12/01/2010] [Accepted: 12/07/2010] [Indexed: 01/16/2023]
|
29
|
Genes for embryo development are packaged in blocks of multivalent chromatin in zebrafish sperm. Genome Res 2011; 21:578-89. [PMID: 21383318 DOI: 10.1101/gr.113167.110] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In mature human sperm, genes of importance for embryo development (i.e., transcription factors) lack DNA methylation and bear nucleosomes with distinctive histone modifications, suggesting the specialized packaging of these developmental genes in the germline. Here, we explored the tractable zebrafish model and found conceptual conservation as well as several new features. Biochemical and mass spectrometric approaches reveal the zebrafish sperm genome packaged in nucleosomes and histone variants (and not protamine), and we find linker histones high and H4K16ac absent, key factors that may contribute to genome condensation. We examined several activating (H3K4me2/3, H3K14ac, H2AFV) and repressing (H3K27me3, H3K36me3, H3K9me3, hypoacetylation) modifications/compositions genome-wide and find developmental genes packaged in large blocks of chromatin with coincident activating and repressing marks and DNA hypomethylation, revealing complex "multivalent" chromatin. Notably, genes that acquire DNA methylation in the soma (muscle) are enriched in transcription factors for alternative cell fates. Remarkably, whereas H3K36me3 is located in the 3' coding region of heavily transcribed genes in somatic cells, H3K36me3 is present in the promoters of "silent" developmental regulators in sperm, suggesting different rules for H3K36me3 in the germline and soma. We also reveal the chromatin patterns of transposons, rDNA, and tDNAs. Finally, high levels of H3K4me3 and H3K14ac in sperm are correlated with genes activated in embryos prior to the mid-blastula transition (MBT), whereas multivalent genes are correlated with activation at or after MBT. Taken together, gene sets with particular functions in the embryo are packaged by distinctive types of complex and often atypical chromatin in sperm.
Collapse
|
30
|
HOCHI S, ABDALLA H, HARA H, HIRABAYASHI M. Challenging Endeavour for Preservation of Freeze-Dried Mammalian Spermatozoa. J Reprod Dev 2011; 57:557-63. [DOI: 10.1262/jrd.11-061o] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Shinichi HOCHI
- Graduate School of Science and Technology, Shinshu University, Nagano 386-8567, Japan
| | - Hany ABDALLA
- Graduate School of Science and Technology, Shinshu University, Nagano 386-8567, Japan
- Faculty of Veterinary Medicine, Zagazig University, Sharkia 44519, Egypt
| | - Hiromasa HARA
- Graduate School of Science and Technology, Shinshu University, Nagano 386-8567, Japan
| | | |
Collapse
|
31
|
Dain L, Auslander R, Dirnfeld M. The effect of paternal age on assisted reproduction outcome. Fertil Steril 2011; 95:1-8. [DOI: 10.1016/j.fertnstert.2010.08.029] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 07/12/2010] [Accepted: 08/13/2010] [Indexed: 11/30/2022]
|
32
|
Finer S, Holland ML, Nanty L, Rakyan VK. The hunt for the epiallele. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2011; 52:1-11. [PMID: 20839222 DOI: 10.1002/em.20590] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Understanding the origin of phenotypic variation remains one of the principle challenges of contemporary biology. Recent genome-wide association studies have identified association between common genetic variants and complex phenotype; however, the minimal effect sizes observed in such studies highlight the potential for other causal factors to be involved in phenotypic variation. The epigenetic state of an organism (or 'epigenome') incorporates a landscape of complex and plastic molecular events that may underlie the 'missing link' that integrates genotype with phenotype. The nature of these processes has been the subject of intense scientific study over the recent years, and characterisation of epigenetic variation, in the form of 'epialleles', is providing fascinating insight into how the genome functions within a range of developmental processes, environments, and in states of health and disease. This review will discuss how and when mammalian epialleles may be generated and their interaction with genetic and environmental factors. We will outline how an epiallele has a variable relationship with phenotype, and how new technologies may be used for their detection and to facilitate an understanding of their contribution to phenotype. Finally, we will consider epialleles in population variation and their teleological role in evolution. variation and their teleological role in evolution.
Collapse
Affiliation(s)
- Sarah Finer
- The Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | | | | | | |
Collapse
|
33
|
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
|
34
|
BARNETOVA I, FULKA H, FULKA, JR J. Epigenetic Characteristics of Paternal Chromatin in Interspecies Zygotes. J Reprod Dev 2010; 56:601-6. [DOI: 10.1262/jrd.09-172a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Irena BARNETOVA
- Institute of Animal Science
- Center for Cell Therapy and Tissue Repair
| | - Helena FULKA
- Institute of Animal Science
- Center for Cell Therapy and Tissue Repair
| | - Josef FULKA, JR
- Institute of Animal Science
- Center for Cell Therapy and Tissue Repair
| |
Collapse
|