Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 3861, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35790717

ABSTRACT

Mammalian pre-implantation embryos accumulate substantial lipids, which are stored in lipid droplets (LDs). Despite the fundamental roles of lipids in many cellular functions, the significance of building-up LDs for the developing embryo remains unclear. Here we report that the accumulation and mobilization of LDs upon implantation are causal in the morphogenesis of the pluripotent epiblast and generation of the pro-amniotic cavity in mouse embryos, a critical step for all subsequent development. We show that the CIDEA protein, found abundantly in adipocytes, enhances lipid storage in blastocysts and pluripotent stem cells by promoting LD enlargement through fusion. The LD-stored lipids are mobilized into lysosomes at the onset of lumenogenesis, but without CIDEA are prematurely degraded by cytosolic lipases. Loss of lipid storage or inactivation of lipophagy leads to the aberrant formation of multiple cavities within disorganised epithelial structures. Thus, our study reveals an unexpected role for LDs in orchestrating tissue remodelling and uncovers underappreciated facets of lipid metabolism in peri-implantation development.


Subject(s)
Lipid Droplets , Lipid Metabolism , Adipocytes/metabolism , Animals , Lipid Droplets/metabolism , Lipid Metabolism/physiology , Lipids , Mammals , Mice , Morphogenesis
2.
Mol Biol Evol ; 37(7): 1986-2001, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32145025

ABSTRACT

Genetic variation in the enzymes that catalyze posttranslational modification of proteins is a potentially important source of phenotypic variation during evolution. Ubiquitination is one such modification that affects turnover of virtually all of the proteins in the cell in addition to roles in signaling and epigenetic regulation. UBE2D3 is a promiscuous E2 enzyme, which acts as an ubiquitin donor for E3 ligases that catalyze ubiquitination of developmentally important proteins. We have used protein sequence comparison of UBE2D3 orthologs to identify a position in the C-terminal α-helical region of UBE2D3 that is occupied by a conserved serine in amniotes and by alanine in anamniote vertebrate and invertebrate lineages. Acquisition of the serine (S138) in the common ancestor to modern amniotes created a phosphorylation site for Aurora B. Phosphorylation of S138 disrupts the structure of UBE2D3 and reduces the level of the protein in mouse embryonic stem cells (ESCs). Substitution of S138 with the anamniote alanine (S138A) increases the level of UBE2D3 in ESCs as well as being a gain of function early embryonic lethal mutation in mice. When mutant S138A ESCs were differentiated into extraembryonic primitive endoderm, levels of the PDGFRα and FGFR1 receptor tyrosine kinases were reduced and primitive endoderm differentiation was compromised. Proximity ligation analysis showed increased interaction between UBE2D3 and the E3 ligase CBL and between CBL and the receptor tyrosine kinases. Our results identify a sequence change that altered the ubiquitination landscape at the base of the amniote lineage with potential effects on amniote biology and evolution.


Subject(s)
Endoderm/enzymology , Evolution, Molecular , Proto-Oncogene Proteins c-cbl/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Vertebrates/genetics , Amino Acid Substitution , Animals , Aurora Kinase B/metabolism , Female , Humans , Mice , Phosphorylation , Receptor Protein-Tyrosine Kinases/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Vertebrates/metabolism
3.
Nat Commun ; 11(1): 1112, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111830

ABSTRACT

Clusters of enhancers, referred as to super-enhancers (SEs), control the expression of cell identity genes. The organisation of these clusters, and how they are remodelled upon developmental transitions remain poorly understood. Here, we report the existence of two types of enhancer units within SEs typified by distinctive CpG methylation dynamics in embryonic stem cells (ESCs). We find that these units are either prone for decommissioning or remain constitutively active in epiblast stem cells (EpiSCs), as further established in the peri-implantation epiblast in vivo. Mechanistically, we show a pivotal role for ESRRB in regulating the activity of ESC-specific enhancer units and propose that the developmentally regulated silencing of ESRRB triggers the selective inactivation of these units within SEs. Our study provides insights into the molecular events that follow the loss of ESRRB binding, and offers a mechanism by which the naive pluripotency transcriptional programme can be partially reset upon embryo implantation.


Subject(s)
CpG Islands , DNA Methylation , Enhancer Elements, Genetic/genetics , Pluripotent Stem Cells/metabolism , Receptors, Estrogen/metabolism , Animals , Gene Expression Regulation, Developmental , Germ Layers/cytology , Mediator Complex/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Protein Binding , RNA Polymerase II/metabolism , Transcription, Genetic
4.
Stem Cell Res Ther ; 10(1): 122, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30999923

ABSTRACT

BACKGROUND: Sox2 is a well-established pluripotent transcription factor that plays an essential role in establishing and maintaining pluripotent stem cells (PSCs). It is also thought to be a linage specifier that governs PSC neural lineage specification upon their exiting the pluripotent state. However, the exact role of SOX2 in human PSCs was still not fully understood. In this study, we studied the role of SOX2 in human embryonic stem cells (hESCs) by gain- and loss-of-function approaches and explored the possible underlying mechanisms. RESULTS: We demonstrate that knockdown of SOX2 induced hESC differentiation to endoderm-like cells, whereas overexpression of SOX2 in hESCs enhanced their pluripotency under self-renewing culture conditions but promoted their neural differentiation upon replacing the culture to non-self-renewal conditions. We show that this culture-dependent dual function of SOX2 was probably attributed to its interaction with different transcription factors predisposed by the culture environments. Whilst SOX2 interacts with OCT4 under self-renewal conditions, we found that, upon neural differentiation, PAX6, a key neural transcription factor, is upregulated and shows interaction with SOX2. The SOX2-PAX6 complex has different gene regulation pattern from that of SOX2-OCT4 complex. CONCLUSIONS: Our work provides direct evidence that SOX2 is necessarily required for hESC pluripotency; however, it can also function as a neural factor, depending on the environmental input. OCT4 and PAX6 might function as key SOX2-interacting partners that determine the function of SOX2 in hESCs.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , PAX6 Transcription Factor/metabolism , SOXB1 Transcription Factors/metabolism , Cell Line , Human Embryonic Stem Cells/cytology , Humans , Neural Stem Cells/cytology
5.
Development ; 144(4): 567-579, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087629

ABSTRACT

Jmjd2 H3K9 demethylases cooperate in promoting mouse embryonic stem cell (ESC) identity. However, little is known about their importance at the exit of ESC pluripotency. Here, we reveal that Jmjd2c facilitates this process by stabilising the assembly of mediator-cohesin complexes at lineage-specific enhancers. Functionally, we show that Jmjd2c is required in ESCs to initiate appropriate gene expression programs upon somatic multi-lineage differentiation. In the absence of Jmjd2c, differentiation is stalled at an early post-implantation epiblast-like stage, while Jmjd2c-knockout ESCs remain capable of forming extra-embryonic endoderm derivatives. Dissection of the underlying molecular basis revealed that Jmjd2c is re-distributed to lineage-specific enhancers during ESC priming for differentiation. Interestingly, Jmjd2c-bound enhancers are co-occupied by the H3K9-methyltransferase G9a (also known as Ehmt2), independently of its H3K9-modifying activity. Loss of Jmjd2c abrogates G9a recruitment and further destabilises loading of the mediator and cohesin components Med1 and Smc1a at newly activated and poised enhancers in ESC-derived epiblast-like cells. These findings unveil Jmjd2c and G9a as novel enhancer-associated factors, and implicate Jmjd2c as a molecular scaffold for the assembly of essential enhancer-protein complexes with an impact on timely gene activation.


Subject(s)
Embryonic Stem Cells/cytology , Enhancer Elements, Genetic , Histone-Lysine N-Methyltransferase/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Animals , Cell Cycle Proteins/physiology , Cell Differentiation , Cell Lineage , Chromosomal Proteins, Non-Histone/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Germ Layers/cytology , Histones/metabolism , Mice , Mice, Knockout , Pluripotent Stem Cells/cytology , Protein Binding , Sequence Analysis, RNA , Cohesins
6.
Sci Rep ; 6: 27139, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27256674

ABSTRACT

The first intercellular differences during mammalian embryogenesis arise in the blastocyst, producing the inner cell mass and the trophectoderm. The trophectoderm is the first extraembryonic tissue and does not contribute to the embryo proper, its differentiation instead forming tissues that sustain embryonic development. Crucial roles in extraembryonic differentiation have been identified for certain transcription factors, but a comprehensive picture of the regulation of this early specification is still lacking. Here, we investigated whether the regulatory mechanisms involved in Cdx2 expression in the blastocyst are also utilized in the postimplantation embryo. We analyzed an enhancer that is regulated through Hippo and Notch in the blastocyst trophectoderm, unexpectedly finding that it is inactive in the extraembryonic structures at postimplantation stages. Further analysis identified other Cdx2 regulatory elements including a stem-cell specific regulatory sequence and an element that drives reporter expression in the trophectoderm, a subset of cells in the extraembryonic region of the postimplantation embryo and in trophoblast stem cells. The cross-comparison in this study of cis-regulatory elements employed in the blastocyst, stem cell populations and the postimplantation embryo provides new insights into early mammalian development and suggests a two-step mechanism in Cdx2 regulation.


Subject(s)
Blastocyst/metabolism , CDX2 Transcription Factor/genetics , Enhancer Elements, Genetic , Fetal Stem Cells/metabolism , Trophoblasts/metabolism , Animals , Blastocyst/cytology , CDX2 Transcription Factor/metabolism , Cell Differentiation , Cells, Cultured , Embryo Implantation , Embryonic Development , Female , Fetal Stem Cells/cytology , Gene Expression Regulation, Developmental , Mice , Transcription Factors/metabolism , Trophoblasts/cytology
8.
Genes Dev ; 26(20): 2286-98, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-23019124

ABSTRACT

Embryonic stem cell (ESC) pluripotency depends on a well-characterized gene regulatory network centered on Oct4, Sox2, and Nanog. In contrast, little is known about the identity of the key coregulators and the mechanisms by which they may potentiate transcription in ESCs. Alongside core transcription factors, the orphan nuclear receptor Esrrb (estrogen-related receptor ß) is vital for the maintenance of ESC identity and furthermore is uniquely associated with the basal transcription machinery. Here, we show that Ncoa3, an essential coactivator, is required to mediate Esrrb function in ESCs. Ncoa3 interacts with Esrrb via its ligand-binding domain and bridges Esrrb to RNA polymerase II complexes. Functionally, Ncoa3 is critical for both the induction and maintenance of pluripotency. Through chromatin immunoprecipitation (ChIP) sequencing and microarray experiments, we further demonstrate that Ncoa3 shares overlapping gene regulatory functions with Esrrb and cooperates genome-wide with the Oct4-Sox2-Nanog circuitry at active enhancers to up-regulate genes involved in self-renewal and pluripotency. We propose an integrated model of transcriptional and coactivator control, mediated by Ncoa3, for the maintenance of ESC self-renewal and somatic cell reprogramming.


Subject(s)
Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Nuclear Receptor Coactivator 3/metabolism , Receptors, Estrogen/metabolism , Animals , COS Cells , Cell Proliferation , Chlorocebus aethiops , Female , Gene Expression Regulation, Developmental , Genome/genetics , HEK293 Cells , Humans , Male , Mice , Receptors, Estrogen/genetics
9.
Genes Dev ; 26(13): 1445-58, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22713603

ABSTRACT

The transcription factors Nanog and Gata6 are critical to specify the epiblast versus primitive endoderm (PrE) lineages. However, little is known about the mechanisms that regulate the protein stability and activity of these factors in the developing embryo. Here we uncover an early developmental function for the Polycomb group member Bmi1 in supporting PrE lineage formation through Gata6 protein stabilization. We show that Bmi1 is enriched in the extraembryonic (endoderm [XEN] and trophectodermal stem [TS]) compartment and repressed by Nanog in pluripotent embryonic stem (ES) cells. In vivo, Bmi1 overlaps with the nascent Gata6 and Nanog protein from the eight-cell stage onward before it preferentially cosegregates with Gata6 in PrE progenitors. Mechanistically, we demonstrate that Bmi1 interacts with Gata6 in a Ring finger-dependent manner to confer protection against Gata6 ubiquitination and proteasomal degradation. A direct role for Bmi1 in cell fate allocation is established by loss-of-function experiments in chimeric embryoid bodies. We thus propose a novel regulatory pathway by which Bmi1 action on Gata6 stability could alter the balance between Gata6 and Nanog protein levels to introduce a bias toward a PrE identity in a cell-autonomous manner.


Subject(s)
Endoderm/metabolism , GATA6 Transcription Factor/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cell Lineage , Endoderm/cytology , GATA6 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Nanog Homeobox Protein , Nuclear Proteins/genetics , Pluripotent Stem Cells/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Transcription, Genetic
10.
Cell Stem Cell ; 10(1): 33-46, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22226354

ABSTRACT

The Polycomb Group (PcG) of chromatin modifiers regulates pluripotency and differentiation. Mammalian genomes encode multiple homologs of the Polycomb repressive complex 1 (PRC1) components, including five orthologs of the Drosophila Polycomb protein (Cbx2, Cbx4, Cbx6, Cbx7, and Cbx8). We have identified Cbx7 as the primary Polycomb ortholog of PRC1 complexes in embryonic stem cells (ESCs). The expression of Cbx7 is downregulated during ESC differentiation, preceding the upregulation of Cbx2, Cbx4, and Cbx8, which are directly repressed by Cbx7. Ectopic expression of Cbx7 inhibits differentiation and X chromosome inactivation and enhances ESC self-renewal. Conversely, Cbx7 knockdown induces differentiation and derepresses lineage-specific markers. In a functional screen, we identified the miR-125 and miR-181 families as regulators of Cbx7 that are induced during ESC differentiation. Ectopic expression of these miRNAs accelerates ESC differentiation via regulation of Cbx7. These observations establish a critical role for Cbx7 and its regulatory miRNAs in determining pluripotency.


Subject(s)
Cell Differentiation/physiology , Down-Regulation/physiology , Embryonic Stem Cells/metabolism , MicroRNAs/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/metabolism , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Cell Line, Tumor , Chromosomes, Human, X/genetics , Chromosomes, Human, X/metabolism , Embryonic Stem Cells/cytology , Humans , Ligases , Mice , MicroRNAs/genetics , Mitochondrial Membrane Transport Proteins , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Repressor Proteins/genetics , Ubiquitin-Protein Ligases , X Chromosome Inactivation/physiology
11.
Development ; 137(15): 2483-92, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20573702

ABSTRACT

Pluripotent cells develop within the inner cell mass of blastocysts, a mosaic of cells surrounded by an extra-embryonic layer, the trophectoderm. We show that a set of somatic lineage regulators (including Hox, Gata and Sox factors) that carry bivalent chromatin enriched in H3K27me3 and H3K4me2 are selectively targeted by Suv39h1-mediated H3K9me3 and de novo DNA methylation in extra-embryonic versus embryonic (pluripotent) lineages, as assessed both in blastocyst-derived stem cells and in vivo. This stably repressed state is linked with a loss of gene priming for transcription through the exclusion of PRC1 (Ring1B) and RNA polymerase II complexes at bivalent, lineage-inappropriate genes upon trophoblast lineage commitment. Collectively, our results suggest a mutually exclusive role for Ring1B and Suv39h1 in regulating distinct chromatin states at key developmental genes and propose a novel mechanism by which lineage specification can be reinforced during early development.


Subject(s)
Chromatin/chemistry , Gene Expression Regulation, Developmental , Methyltransferases/physiology , Repressor Proteins/physiology , Animals , Blastocyst , Cell Lineage , Chromatin/metabolism , DNA Methylation , Gene Expression Profiling , Gene Silencing , Methyltransferases/metabolism , Mice , Models, Biological , Polycomb Repressive Complex 1 , RNA Interference , RNA Polymerase II/metabolism , Repressor Proteins/metabolism , Trophoblasts/metabolism , Ubiquitin-Protein Ligases
12.
Epigenetics Chromatin ; 3: 1, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-20157423

ABSTRACT

BACKGROUND: During early mouse development, two extra-embryonic lineages form alongside the future embryo: the trophectoderm (TE) and the primitive endoderm (PrE). Epigenetic changes known to take place during these early stages include changes in DNA methylation and modified histones, as well as dynamic changes in gene expression. RESULTS: In order to understand the role and extent of chromatin-based changes for lineage commitment within the embryo, we examined the epigenetic profiles of mouse embryonic stem (ES), trophectoderm stem (TS) and extra-embryonic endoderm (XEN) stem cell lines that were derived from the inner cell mass (ICM), TE and PrE, respectively. As an initial indicator of the chromatin state, we assessed the replication timing of a cohort of genes in each cell type, based on data that expressed genes and acetylated chromatin domains, generally, replicate early in S-phase, whereas some silent genes, hypoacetylated or condensed chromatin tend to replicate later. We found that many lineage-specific genes replicate early in ES, TS and XEN cells, which was consistent with a broadly 'accessible' chromatin that was reported previously for multiple ES cell lines. Close inspection of these profiles revealed differences between ES, TS and XEN cells that were consistent with their differing lineage affiliations and developmental potential. A comparative analysis of modified histones at the promoters of individual genes showed that in TS and ES cells many lineage-specific regulator genes are co-marked with modifications associated with active (H4ac, H3K4me2, H3K9ac) and repressive (H3K27me3) chromatin. However, in XEN cells several of these genes were marked solely by repressive modifications (such as H3K27me3, H4K20me3). Consistent with TS and XEN having a restricted developmental potential, we show that these cells selectively reprogramme somatic cells to induce the de novo expression of genes associated with extraembryonic differentiation. CONCLUSIONS: These data provide evidence that the diversification of defined embryonic and extra-embryonic lineages is accompanied by chromatin remodelling at specific loci. Stem cell lines from the ICM, TE and PrE can each dominantly reprogramme somatic cells but reset gene expression differently, reflecting their separate lineage identities and increasingly restricted developmental potentials.

13.
Genes Dev ; 23(18): 2134-9, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19696146

ABSTRACT

Somatic cells can be reprogrammed into induced pluripotent stem (iPS) cells by overexpressing combinations of factors such as Oct4, Sox2, Klf4, and c-Myc. Reprogramming is slow and stochastic, suggesting the existence of barriers limiting its efficiency. Here we identify senescence as one such barrier. Expression of the four reprogramming factors triggers senescence by up-regulating p53, p16(INK4a), and p21(CIP1). Induction of DNA damage response and chromatin remodeling of the INK4a/ARF locus are two of the mechanisms behind senescence induction. Crucially, ablation of different senescence effectors improves the efficiency of reprogramming, suggesting novel strategies for maximizing the generation of iPS cells.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming , Cellular Senescence/genetics , Gene Expression Regulation , Pluripotent Stem Cells/cytology , Animals , Cell Line , Humans , Kruppel-Like Factor 4
14.
Cell Cycle ; 6(19): 2393-8, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17938583

ABSTRACT

The timing of DNA replication generally correlates with transcription, gene density and sequence composition. How is the timing affected if a genomic region has a combination of features that individually correlate with either early or late replication? The major histocompatibility complex (MHC) class II region is an AT-rich isochore that would be expected to replicate late, but it also contains coordinately regulated genes that are highly expressed in antigen-presenting cells and are strongly inducible in other cell types. Using cytological and biochemical assays, we find that the entire MHC replicates within the first half of S-phase, and that the class II region replicates slightly later than the adjacent regions irrespective of gene expression. These data suggest that despite AT-richness, an early-to-middle replication time in the class II region is defined by an open chromatin conformation that allows rapid transcriptional activation as a defence against pathogens.


Subject(s)
DNA Replication Timing , Genes, MHC Class II , Histocompatibility Antigens Class II/metabolism , Interferon-gamma/metabolism , Animals , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , Histocompatibility Antigens Class II/genetics , Transcription, Genetic
15.
Genome Biol ; 8(8): R169, 2007.
Article in English | MEDLINE | ID: mdl-17705870

ABSTRACT

BACKGROUND: The time of locus replication during S-phase is tightly regulated and correlates with chromatin state. Embryonic stem (ES) cells have an unusual chromatin profile where many developmental regulator genes that are not yet expressed are marked by both active and repressive histone modifications. This poised or bivalent state is also characterized by locus replication in early S-phase in ES cells, while replication timing is delayed in cells with restricted developmental options. RESULTS: Here we used a panel of mutant mouse ES cell lines lacking important chromatin modifiers to dissect the relationship between chromatin structure and replication timing. We show that temporal control of satellite DNA replication is sensitive to loss of a variety of chromatin modifiers, including Mll, Eed, Dnmt1, Suv39h1/h2 and Dicer. The replication times of many single copy loci, including a 5 Mb contiguous region surrounding the Rex1 gene, were retained in chromatin modifier mutant ES cells, although a subset of loci were affected. CONCLUSION: This analysis demonstrates the importance of chromatin modifiers for maintaining correct replication of satellite sequences in pluripotent ES cells and highlights the sensitivity of some single copy loci to the influence of chromatin modifiers. Abundant histone acetylation is shown to correlate well with early replication. Surprisingly, loss of DNA methylation or histone methylation was tolerated by many loci, suggesting that these modifications may be less influential for the timing of euchromatin replication.


Subject(s)
Chromatin/metabolism , DNA Replication , DNA, Satellite/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Acetylation , Animals , Cell Line , DNA Methylation , Histones/metabolism , Mice , Mutation , S Phase/genetics
16.
J Biol Chem ; 282(16): 12194-200, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17311920

ABSTRACT

A large fraction of the animal genome is maintained in a transcriptionally repressed state throughout development. By generating viable Dnmt1(-)(/)(-) mouse cells we have been able to study the effect of DNA methylation on both gene expression and chromatin structure. Our results confirm that the underlying methylation pattern has a profound effect on histone acetylation and is the major effector of me-H3(K4) in the animal genome. We demonstrate that many methylated genes are subject to additional repression mechanisms that also impact on histone acetylation, and the data suggest that late replication timing may play an important role in this process.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methylation , Gene Expression Regulation , Acetylation , Animals , Chromatin/chemistry , CpG Islands , DNA/chemistry , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Replication , Epigenesis, Genetic , Fibroblasts/metabolism , Histones/chemistry , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis
17.
Nat Cell Biol ; 8(5): 532-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16570078

ABSTRACT

Epigenetic genome modifications are thought to be important for specifying the lineage and developmental stage of cells within a multicellular organism. Here, we show that the epigenetic profile of pluripotent embryonic stem cells (ES) is distinct from that of embryonic carcinoma cells, haematopoietic stem cells (HSC) and their differentiated progeny. Silent, lineage-specific genes replicated earlier in pluripotent cells than in tissue-specific stem cells or differentiated cells and had unexpectedly high levels of acetylated H3K9 and methylated H3K4. Unusually, in ES cells these markers of open chromatin were also combined with H3K27 trimethylation at some non-expressed genes. Thus, pluripotency of ES cells is characterized by a specific epigenetic profile where lineage-specific genes may be accessible but, if so, carry repressive H3K27 trimethylation modifications. H3K27 methylation is functionally important for preventing expression of these genes in ES cells as premature expression occurs in embryonic ectoderm development (Eed)-deficient ES cells. Our data suggest that lineage-specific genes are primed for expression in ES cells but are held in check by opposing chromatin modifications.


Subject(s)
Chromatin/genetics , Pluripotent Stem Cells/metabolism , Animals , Carcinoma/genetics , Cell Line , Cells, Cultured , DNA Replication Timing/genetics , Down-Regulation/genetics , Epigenesis, Genetic , Gene Expression Profiling , Genetic Markers , Hematopoietic Stem Cells/metabolism , Mice , Multipotent Stem Cells/metabolism , Polycomb Repressive Complex 2 , Repressor Proteins/metabolism , T-Lymphocytes/metabolism
18.
J Cell Sci ; 119(Pt 1): 132-40, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16371653

ABSTRACT

Determining how genes are epigenetically regulated to ensure their correct spatial and temporal expression during development is key to our understanding of cell lineage commitment. Here we examined epigenetic changes at an important proneural regulator gene Mash1 (Ascl1), as embryonic stem (ES) cells commit to the neural lineage. In ES cells where the Mash1 gene is transcriptionally repressed, the locus replicated late in S phase and was preferentially positioned at the nuclear periphery with other late-replicating genes (Neurod, Sprr2a). This peripheral location was coupled with low levels of histone H3K9 acetylation at the Mash1 promoter and enhanced H3K27 methylation but surprisingly location was not affected by removal of the Ezh2/Eed HMTase complex or several other chromatin-silencing candidates (G9a, SuV39h-1, Dnmt-1, Dnmt-3a and Dnmt-3b). Upon neural induction however, Mash1 transcription was upregulated (>100-fold), switched its time of replication from late to early in S phase and relocated towards the interior of the nucleus. This spatial repositioning was selective for neural commitment because Mash1 was peripheral in ES-derived mesoderm and other non-neural cell types. A bidirectional analysis of replication timing across a 2 Mb region flanking the Mash1 locus showed that chromatin changes were focused at Mash1. These results suggest that Mash1 is regulated by changes in chromatin structure and location and implicate the nuclear periphery as an important environment for maintaining the undifferentiated state of ES cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Chromatin/metabolism , Embryonic Induction/physiology , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Neurons/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Line , Cell Lineage , Cell Nucleus/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Mice , Mice, Inbred C57BL , Neurons/cytology , Nucleic Acid Conformation , Protein Methyltransferases , Transcription, Genetic
19.
Nat Protoc ; 1(4): 2171-7, 2006.
Article in English | MEDLINE | ID: mdl-17487209

ABSTRACT

Profiling chromatin in a particular cell type provides a valuable 'signature' for cell identity and developmental stage. One approach has been to assay and use the timing of DNA replication across a panel of loci as an indicator of chromatin structure. This epigenetic profiling used on pluripotent embryonic stem (ES) cells has reliably distinguished them from cells that have a more restricted lineage potential. Thus, such an approach may become increasingly useful for understanding the molecular basis of pluripotency and lineage induction, especially in the context of stem-cell therapy. Here I describe in detail the DNA replication timing method, whereby unsynchronized cell populations are pulse-labeled with 5-bromo-2'-deoxyuridine (BrdU), fractionated according to cell-cycle stage and the abundance of candidate sequences within newly replicated DNA is determined by PCR. This robust protocol has been used consistently by several laboratories and might offer some advantages over conventional transcription-based profiling for characterizing cell populations. The procedure requires 3-4 d to complete.


Subject(s)
DNA Replication Timing , S Phase , Animals , DNA/chemistry , Flow Cytometry , Humans , Mice
20.
Cell Cycle ; 3(12): 1645-50, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15611653

ABSTRACT

Mammalian embryonic stem (ES) cells can either self-renew or generate progenitor cells that have a more restricted developmental potential. This provides an important model system to ask how pluripotency, cell commitment and differentiation are regulated at the level of chromatin-based changes that distinguish stem cells from their differentiated progeny. Here we show that the differentiation of ES cells to neural progenitors results in dynamic changes in the epigenetic status of multiple genes that encode transcription factors critical for early embryonic development or lineage specification. In particular, we demonstrate that DNA replication at a subset of neural-associated genes including Pax3, Pax6, Irx3, Nkx2.9 and Mash1 is advanced upon neural induction, consistent with increased locus accessibility. Conversely, many ES-associated genes including Oct4, Nanog, Utf1, Foxd3, Cripto and Rex1 that replicate early in ES cells switch their replication timing to later in S-phase in response to differentiation. Detailed analysis of the Rex1 locus reveals that delayed replication extends to a 2.8 Mb region surrounding the gene and is associated with substantial reductions in the level of histone H3K9 and H4 acetylation at the promoter. These results show that loss of pluripotency (and lineage choice) is associated with extensive and predictable changes in the replication timing of key regulator genes.


Subject(s)
DNA Replication Timing , Gene Expression Regulation, Developmental , Genes, Regulator/genetics , Nervous System/embryology , Neurons/cytology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Base Composition , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Flow Cytometry , Genome , Histones/metabolism , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/drug effects , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...