Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 10: 1014498, 2022.
Article in English | MEDLINE | ID: mdl-36299478

ABSTRACT

Cellular metabolism is a complex network of biochemical reactions fueling development with energy and biomass; however, it can also shape the cellular epigenome. Indeed, some intermediates of metabolic reactions exert a non-canonical function by acting as co-factors, substrates or inhibitors of chromatin modifying enzymes. Therefore, fluctuating availability of such molecules has the potential to regulate the epigenetic landscape. Thanks to this functional coupling, chromatin can act as a sensor of metabolic changes and thus impact cell fate. Growing evidence suggest that both metabolic and epigenetic reprogramming are crucial for ensuring a successful embryo development from the zygote until gastrulation. In this review, we provide an overview of the complex relationship between metabolism and epigenetics in regulating the early stages of mammalian embryo development. We report on recent breakthroughs in uncovering the non-canonical functions of metabolism especially when re-localized to the nucleus. In addition, we identify the challenges and outline future perspectives to advance the novel field of epi-metabolomics especially in the context of early development.

2.
EMBO Rep ; 22(3): e51989, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33605056

ABSTRACT

During X chromosome inactivation (XCI), in female placental mammals, gene silencing is initiated by the Xist long non-coding RNA. Xist accumulation at the X leads to enrichment of specific chromatin marks, including PRC2-dependent H3K27me3 and SETD8-dependent H4K20me1. However, the dynamics of this process in relation to Xist RNA accumulation remains unknown as is the involvement of H4K20me1 in initiating gene silencing. To follow XCI dynamics in living cells, we developed a genetically encoded, H3K27me3-specific intracellular antibody or H3K27me3-mintbody. By combining live-cell imaging of H3K27me3, H4K20me1, the X chromosome and Xist RNA, with ChIP-seq analysis we uncover concurrent accumulation of both marks during XCI, albeit with distinct genomic distributions. Furthermore, using a Xist B and C repeat mutant, which still shows gene silencing on the X but not H3K27me3 deposition, we also find a complete lack of H4K20me1 enrichment. This demonstrates that H4K20me1 is dispensable for the initiation of gene silencing, although it may have a role in the chromatin compaction that characterises facultative heterochromatin.


Subject(s)
Histones , RNA, Long Noncoding , Animals , Female , Gene Silencing , Histones/genetics , Histones/metabolism , Placenta/metabolism , Pregnancy , RNA, Long Noncoding/genetics , X Chromosome/genetics , X Chromosome Inactivation/genetics
3.
Cells ; 9(12)2020 12 17.
Article in English | MEDLINE | ID: mdl-33348832

ABSTRACT

Dosage compensation between the sexes results in one X chromosome being inactivated during female mammalian development. Chromosome-wide transcriptional silencing from the inactive X chromosome (Xi) in mammalian cells is erased in a process termed X-chromosome reactivation (XCR), which has emerged as a paradigm for studying the reversal of chromatin silencing. XCR is linked with germline development and induction of naive pluripotency in the epiblast, and also takes place upon reprogramming somatic cells to induced pluripotency. XCR depends on silencing of the long non-coding RNA (lncRNA) X inactive specific transcript (Xist) and is linked with the erasure of chromatin silencing. Over the past years, the advent of transcriptomics and epigenomics has provided new insights into the transcriptional and chromatin dynamics with which XCR takes place. However, multiple questions remain unanswered about how chromatin and transcription related processes enable XCR. Here, we review recent work on establishing the transcriptional and chromatin kinetics of XCR, as well as discuss a model by which transcription factors mediate XCR not only via Xist repression, but also by direct targeting of X-linked genes.


Subject(s)
Cellular Reprogramming , X Chromosome/physiology , Animals , Cell Differentiation , Chromatin/metabolism , Embryonic Development/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
4.
Annu Rev Biochem ; 89: 255-282, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32259458

ABSTRACT

Facultative heterochromatin (fHC) concerns the developmentally regulated heterochromatinization of different regions of the genome and, in the case of the mammalian X chromosome and imprinted loci, of only one allele of a homologous pair. The formation of fHC participates in the timely repression of genes, by resisting strong trans activators. In this review, we discuss the molecular mechanisms underlying the establishment and maintenance of fHC in mammals using a mouse model. We focus on X-chromosome inactivation (XCI) as a paradigm for fHC but also relate it to genomic imprinting and homeobox (Hox) gene cluster repression. A vital role for noncoding transcription and/or transcripts emerges as the general principle of triggering XCI and canonical imprinting. However, other types of fHC are established through an unknown mechanism, independent of noncoding transcription (Hox clusters and noncanonical imprinting). We also extensively discuss polycomb-group repressive complexes (PRCs), which frequently play a vital role in fHC maintenance.


Subject(s)
Gene Expression Regulation, Developmental , Genomic Imprinting , Heterochromatin/metabolism , Polycomb-Group Proteins/genetics , X Chromosome Inactivation , X Chromosome/metabolism , Animals , Chromatin Assembly and Disassembly , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Female , Gene Silencing , Heterochromatin/chemistry , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Male , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Polycomb-Group Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Spermatozoa/cytology , Spermatozoa/growth & development , Spermatozoa/metabolism , X Chromosome/chemistry
5.
Nature ; 578(7795): 455-460, 2020 02.
Article in English | MEDLINE | ID: mdl-32025035

ABSTRACT

Xist represents a paradigm for the function of long non-coding RNA in epigenetic regulation, although how it mediates X-chromosome inactivation (XCI) remains largely unexplained. Several proteins that bind to Xist RNA have recently been identified, including the transcriptional repressor SPEN1-3, the loss of which has been associated with deficient XCI at multiple loci2-6. Here we show in mice that SPEN is a key orchestrator of XCI in vivo and we elucidate its mechanism of action. We show that SPEN is essential for initiating gene silencing on the X chromosome in preimplantation mouse embryos and in embryonic stem cells. SPEN is dispensable for maintenance of XCI in neural progenitors, although it significantly decreases the expression of genes that escape XCI. We show that SPEN is immediately recruited to the X chromosome upon the upregulation of Xist, and is targeted to enhancers and promoters of active genes. SPEN rapidly disengages from chromatin upon gene silencing, suggesting that active transcription is required to tether SPEN to chromatin. We define the SPOC domain as a major effector of the gene-silencing function of SPEN, and show that tethering SPOC to Xist RNA is sufficient to mediate gene silencing. We identify the protein partners of SPOC, including NCoR/SMRT, the m6A RNA methylation machinery, the NuRD complex, RNA polymerase II and factors involved in the regulation of transcription initiation and elongation. We propose that SPEN acts as a molecular integrator for the initiation of XCI, bridging Xist RNA with the transcription machinery-as well as with nucleosome remodellers and histone deacetylases-at active enhancers and promoters.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Silencing , RNA-Binding Proteins/metabolism , Transcription, Genetic , X Chromosome Inactivation/genetics , X Chromosome/genetics , Animals , Blastocyst/cytology , Blastocyst/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/chemistry , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic/genetics , Female , Histone Deacetylases/metabolism , Male , Methylation , Mice , Promoter Regions, Genetic/genetics , Protein Domains , RNA, Long Noncoding/genetics , RNA-Binding Proteins/chemistry
6.
Curr Protoc Stem Cell Biol ; 52(1): e105, 2020 03.
Article in English | MEDLINE | ID: mdl-31971672

ABSTRACT

The complex program of mouse development entails specification of the embryonic epiblast (Epi) as well as the extra-embryonic trophectoderm (TE) and primitive endoderm (PrE). These three lineages of mouse blastocyst can be modeled in vitro using stem cells derived from primary tissues. In these cultures, cells self-renew while retaining their developmental potential if put back into a developing embryo. Indeed, embryonic stem cells (ESC), when injected into a blastocyst, readily contribute to all embryonic lineages. Similarly, trophoblast stem cells (TSCs) will give rise to all TE-derived trophoblast lineages, and extraembryonic endoderm cells (XEN) will contribute to the PrE-derived yolk sack. These model systems are a powerful tool to study early development, lineage specification, and placenta formation. Only recently reproducible and chemically defined culture systems of these cells have been described. This overview discusses such novel methods for culturing ESC/TSC/XEN, as well as their molecular signatures and developmental potential. Recent strides in expanding the developmental potential of stem cells as well as achieving models more reminiscent of their in vivo counterparts are discussed. Finally, such in vitro stem cells can self-assemble into structures resembling embryos when used in novel 3D-culture systems. This article discusses the strengths and limitations of such "synthetic embryos" in studying developmental processes. © 2020 by John Wiley & Sons, Inc.


Subject(s)
Blastocyst/cytology , Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Models, Biological , Animals , Cell Lineage , Embryonic Development , Mice , Trophoblasts/cytology
7.
Nat Genet ; 51(6): 1024-1034, 2019 06.
Article in English | MEDLINE | ID: mdl-31133748

ABSTRACT

The mouse X-inactivation center (Xic) locus represents a powerful model for understanding the links between genome architecture and gene regulation, with the non-coding genes Xist and Tsix showing opposite developmental expression patterns while being organized as an overlapping sense/antisense unit. The Xic is organized into two topologically associating domains (TADs) but the role of this architecture in orchestrating cis-regulatory information remains elusive. To explore this, we generated genomic inversions that swap the Xist/Tsix transcriptional unit and place their promoters in each other's TAD. We found that this led to a switch in their expression dynamics: Xist became precociously and ectopically upregulated, both in male and female pluripotent cells, while Tsix expression aberrantly persisted during differentiation. The topological partitioning of the Xic is thus critical to ensure proper developmental timing of X inactivation. Our study illustrates how the genomic architecture of cis-regulatory landscapes can affect the regulation of mammalian developmental processes.


Subject(s)
Gene Expression Regulation, Developmental , RNA, Long Noncoding/genetics , X Chromosome Inactivation , Animals , Cell Differentiation/genetics , Ectopic Gene Expression , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gene Silencing , Genetic Loci , Male , Mice , Models, Biological , Promoter Regions, Genetic , Sequence Inversion , Transcription, Genetic
8.
Elife ; 72018 05 10.
Article in English | MEDLINE | ID: mdl-29745895

ABSTRACT

Early mouse development is regulated and accompanied by dynamic changes in chromatin modifications, including G9a-mediated histone H3 lysine 9 dimethylation (H3K9me2). Previously, we provided insights into its role in post-implantation development (Zylicz et al., 2015). Here we explore the impact of depleting the maternally inherited G9a in oocytes on development shortly after fertilisation. We show that G9a accumulates typically at 4 to 8 cell stage to promote timely repression of a subset of 4 cell stage-specific genes. Loss of maternal inheritance of G9a disrupts the gene regulatory network resulting in developmental delay and destabilisation of inner cell mass lineages by the late blastocyst stage. Our results indicate a vital role of this maternally inherited epigenetic regulator in creating conducive conditions for developmental progression and on cell fate choices.


Subject(s)
Blastocyst/physiology , Cell Differentiation , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Oocytes/physiology , Animals , Gene Regulatory Networks , Mice
9.
Elife ; 62017 03 21.
Article in English | MEDLINE | ID: mdl-28323615

ABSTRACT

The maternal-to-zygotic transition (MZT) marks the period when the embryonic genome is activated and acquires control of development. Maternally inherited factors play a key role in this critical developmental process, which occurs at the 2-cell stage in mice. We investigated the function of the maternally inherited factor Stella (encoded by Dppa3) using single-cell/embryo approaches. We show that loss of maternal Stella results in widespread transcriptional mis-regulation and a partial failure of MZT. Strikingly, activation of endogenous retroviruses (ERVs) is significantly impaired in Stella maternal/zygotic knockout embryos, which in turn leads to a failure to upregulate chimeric transcripts. Amongst ERVs, MuERV-L activation is particularly affected by the absence of Stella, and direct in vivo knockdown of MuERV-L impacts the developmental potential of the embryo. We propose that Stella is involved in ensuring activation of ERVs, which themselves play a potentially key role during early development, either directly or through influencing embryonic gene expression.


Subject(s)
Cell Differentiation , Endogenous Retroviruses/genetics , Gene Expression Regulation, Developmental , Repressor Proteins/metabolism , Zygote/physiology , Animals , Chromosomal Proteins, Non-Histone , Mice
10.
Nature ; 529(7586): 403-407, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26751055

ABSTRACT

Nanog, a core pluripotency factor in the inner cell mass of blastocysts, is also expressed in unipotent primordial germ cells (PGCs) in mice, where its precise role is yet unclear. We investigated this in an in vitro model, in which naive pluripotent embryonic stem (ES) cells cultured in basic fibroblast growth factor (bFGF) and activin A develop as epiblast-like cells (EpiLCs) and gain competence for a PGC-like fate. Consequently, bone morphogenetic protein 4 (BMP4), or ectopic expression of key germline transcription factors Prdm1, Prdm14 and Tfap2c, directly induce PGC-like cells (PGCLCs) in EpiLCs, but not in ES cells. Here we report an unexpected discovery that Nanog alone can induce PGCLCs in EpiLCs, independently of BMP4. We propose that after the dissolution of the naive ES-cell pluripotency network during establishment of EpiLCs, the epigenome is reset for cell fate determination. Indeed, we found genome-wide changes in NANOG-binding patterns between ES cells and EpiLCs, indicating epigenetic resetting of regulatory elements. Accordingly, we show that NANOG can bind and activate enhancers of Prdm1 and Prdm14 in EpiLCs in vitro; BLIMP1 (encoded by Prdm1) then directly induces Tfap2c. Furthermore, while SOX2 and NANOG promote the pluripotent state in ES cells, they show contrasting roles in EpiLCs, as Sox2 specifically represses PGCLC induction by Nanog. This study demonstrates a broadly applicable mechanistic principle for how cells acquire competence for cell fate determination, resulting in the context-dependent roles of key transcription factors during development.


Subject(s)
Enhancer Elements, Genetic/genetics , Germ Cells/cytology , Germ Cells/metabolism , Germ Layers/cytology , Homeodomain Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Transcription Factors/genetics , Activins/pharmacology , Animals , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins , Epigenesis, Genetic , Female , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Developmental , Genome/genetics , Germ Layers/drug effects , Germ Layers/metabolism , Homeodomain Proteins/antagonists & inhibitors , Male , Mice , Mouse Embryonic Stem Cells/drug effects , Nanog Homeobox Protein , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Positive Regulatory Domain I-Binding Factor 1 , Protein Binding , RNA-Binding Proteins , SOXB1 Transcription Factors/metabolism , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Transcription Factors/metabolism
11.
Elife ; 42015 Nov 09.
Article in English | MEDLINE | ID: mdl-26551560

ABSTRACT

Early mouse development is accompanied by dynamic changes in chromatin modifications, including G9a-mediated histone H3 lysine 9 dimethylation (H3K9me2), which is essential for embryonic development. Here we show that genome-wide accumulation of H3K9me2 is crucial for postimplantation development, and coincides with redistribution of enhancer of zeste homolog 2 (EZH2)-dependent histone H3 lysine 27 trimethylation (H3K27me3). Loss of G9a or EZH2 results in upregulation of distinct gene sets involved in cell cycle regulation, germline development and embryogenesis. Notably, the H3K9me2 modification extends to active enhancer elements where it promotes developmentally-linked gene silencing and directly marks promoters and gene bodies. This epigenetic mechanism is important for priming gene regulatory networks for critical cell fate decisions in rapidly proliferating postimplantation epiblast cells.


Subject(s)
Chromatin/metabolism , Embryonic Development , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Animals , Enhancer of Zeste Homolog 2 Protein , Histones/metabolism , Methylation , Mice , Polycomb Repressive Complex 2/metabolism , Protein Processing, Post-Translational
12.
Mol Cell ; 56(4): 564-79, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25457166

ABSTRACT

Primordial germ cells (PGCs) and preimplantation embryos undergo epigenetic reprogramming, which includes comprehensive DNA demethylation. We found that PRMT5, an arginine methyltransferase, translocates from the cytoplasm to the nucleus during this process. Here we show that conditional loss of PRMT5 in early PGCs causes complete male and female sterility, preceded by the upregulation of LINE1 and IAP transposons as well as activation of a DNA damage response. Similarly, loss of maternal-zygotic PRMT5 also leads to IAP upregulation. PRMT5 is necessary for the repressive H2A/H4R3me2s chromatin modification on LINE1 and IAP transposons in PGCs, directly implicating this modification in transposon silencing during DNA hypomethylation. PRMT5 translocates back to the cytoplasm subsequently, to participate in the previously described PIWI-interacting RNA (piRNA) pathway that promotes transposon silencing via de novo DNA remethylation. Thus, PRMT5 is directly involved in genome defense during preimplantation development and in PGCs at the time of global DNA demethylation.


Subject(s)
Blastocyst/enzymology , DNA Methylation , Genomic Instability , Ovum/enzymology , Protein Methyltransferases/physiology , Spermatozoa/enzymology , Animals , Apoptosis , Blastocyst/cytology , Cells, Cultured , DNA Damage , DNA Transposable Elements , Embryonic Development , Embryonic Stem Cells/enzymology , Female , Histones/metabolism , Male , Mice, Transgenic , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases
13.
Science ; 339(6118): 448-52, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23223451

ABSTRACT

Mouse primordial germ cells (PGCs) undergo sequential epigenetic changes and genome-wide DNA demethylation to reset the epigenome for totipotency. Here, we demonstrate that erasure of CpG methylation (5mC) in PGCs occurs via conversion to 5-hydroxymethylcytosine (5hmC), driven by high levels of TET1 and TET2. Global conversion to 5hmC initiates asynchronously among PGCs at embryonic day (E) 9.5 to E10.5 and accounts for the unique process of imprint erasure. Mechanistically, 5hmC enrichment is followed by its protracted decline thereafter at a rate consistent with replication-coupled dilution. The conversion to 5hmC is an important component of parallel redundant systems that drive comprehensive reprogramming in PGCs. Nonetheless, we identify rare regulatory elements that escape systematic DNA demethylation in PGCs, providing a potential mechanistic basis for transgenerational epigenetic inheritance.


Subject(s)
Cytosine/analogs & derivatives , DNA Methylation , Embryo, Mammalian/metabolism , Epigenesis, Genetic , Genomic Imprinting , Germ Cells/metabolism , 5-Methylcytosine/metabolism , Animals , CpG Islands , Cytosine/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Embryonic Development , Female , Germ Layers/cytology , Male , Mice , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/genetics
14.
Trends Genet ; 28(4): 164-74, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22386917

ABSTRACT

Germ cells possess the extraordinary and unique capacity to give rise to a new organism and create an enduring link between all generations. To acquire this property, primordial germ cells (PGCs) transit through an unprecedented programme of sequential epigenetic events that culminates in an epigenomic basal state that is the foundation of totipotency. This process is underpinned by genome-wide DNA demethylation, which may occur through several overlapping pathways, including conversion to 5-hydroxymethylcytosine. We propose that the epigenetic programme in PGCs operates through multiple parallel mechanisms to ensure robustness at the level of individual cells while also being flexible through functional redundancy to guarantee high fidelity of the process. Gaining a better understanding of the molecular mechanisms that direct epigenetic reprogramming in PGCs will enhance our ability to manipulate epigenetic memory, cell-fate decisions and applications in regenerative medicine.


Subject(s)
Cellular Reprogramming , Epigenesis, Genetic , Germ Cells/metabolism , Animals , Cell Lineage , DNA Methylation , Germ Cells/cytology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...