Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Genes Dev ; 29(23): 2435-48, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26584622

ABSTRACT

Elf5 is a transcription factor with pivotal roles in the trophoblast compartment, where it reinforces a trophoblast stem cell (TSC)-specific transcriptional circuit. However, Elf5 is also present in differentiating trophoblast cells that have ceased to express other TSC genes such as Cdx2 and Eomes. In the present study, we aimed to elucidate the context-dependent role of Elf5 at the interface between TSC self-renewal and the onset of differentiation. We demonstrate that precise levels of Elf5 are critical for normal expansion of the TSC compartment and embryonic survival, as Elf5 overexpression triggers precocious trophoblast differentiation. Through integration of protein interactome, transcriptome, and genome-wide chromatin immunoprecipitation data, we reveal that this abundance-dependent function is mediated through a shift in preferred Elf5-binding partners; in TSCs, Elf5 interaction with Eomes recruits Tfap2c to triply occupied sites at TSC-specific genes, driving their expression. In contrast, the Elf5 and Tfap2c interaction becomes predominant as their protein levels increase. This triggers binding to double- and single-occupancy sites that harbor the cognate Tfap2c motif, causing activation of the associated differentiation-promoting genes. These data place Elf5 at the center of a stoichiometry-sensitive transcriptional network, where it acts as a molecular switch governing the balance between TSC proliferation and differentiation.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Regulatory Networks , Transcription Factors/genetics , Transcription Factors/metabolism , Trophoblasts/cytology , Animals , Cell Differentiation/genetics , Cell Line , Cell Self Renewal/genetics , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Developmental/genetics , Mice , Protein Binding , Protein Interaction Domains and Motifs , Transcription Factors/chemistry , Trophoblasts/metabolism
2.
Genes Dev ; 25(16): 1702-15, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21852535

ABSTRACT

The noncoding Tsix RNA is an antisense repressor of Xist and regulates X inactivation in mice. Tsix is essential for preventing the inactivation of the maternally inherited X chromosome in extraembryonic lineages where imprinted X-chromosome inactivation (XCI) occurs. Here we establish an inducible Tsix expression system for investigating Tsix function in development. We show that Tsix has a clear functional window in extraembryonic development. Within this window, Tsix can repress Xist, which is accompanied by DNA methylation of the Xist promoter. As a consequence of Xist repression, reactivation of the inactive X chromosome (Xi) is widely observed. In the parietal endoderm, Tsix represses Xist and causes reactivation of an Xi-linked GFP transgene throughout development, whereas Tsix progressively loses its Xist-repressing function from embryonic day 9.5 (E9.5) onward in trophoblast giant cells and spongiotrophoblast, suggesting that Tsix function depends on a lineage-specific environment. Our data also demonstrate that the maintenance of imprinted XCI requires Xist expression in specific extraembryonic tissues throughout development. This finding shows that reversible XCI is not exclusive to pluripotent cells, and that in some lineages cell differentiation is not accompanied by a stabilization of the Xi.


Subject(s)
RNA, Untranslated/genetics , X Chromosome Inactivation/genetics , X Chromosome/genetics , Animals , Cell Lineage/genetics , DNA Methylation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Situ Hybridization, Fluorescence , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Placenta/cytology , Placenta/embryology , Placenta/metabolism , Pregnancy , RNA, Long Noncoding , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trophoblasts/metabolism
3.
Nucleic Acids Res ; 42(14): 8914-27, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25034692

ABSTRACT

Embryonic stem (ES) cells are in a dynamic equilibrium of distinct functional states, characterized by the heterogeneous expression of critical pluripotency factors and regulated by a spectrum of reversible histone modifications. Maintenance of this equilibrium is a hallmark of pluripotency. Here we find that the ADP-ribosyltransferases Parp1 and Parp7 play a critical role in safeguarding this state by occupying key pluripotency genes, notably Nanog, Pou5f1, Sox2, Stella, Tet1 and Zfp42, thereby protecting them from progressive epigenetic repression. In the absence of either Parp1 or Parp7, or upon inhibition of the ADP-ribosylating activity, ES cells exhibit a decrease in ground state pluripotency as they cannot maintain the typical heterogeneity characteristic of the metastable state. As a consequence, they display a higher propensity to differentiate. These findings place Parp1 and Parp7 at the genetic-epigenetic interface of pluripotency networks, fine-tuning the transcriptional heterogeneity and thereby determining the developmental plasticity of ES cells.


Subject(s)
ADP Ribose Transferases/physiology , Embryonic Stem Cells/enzymology , Pluripotent Stem Cells/enzymology , Poly(ADP-ribose) Polymerases/physiology , ADP Ribose Transferases/genetics , ADP Ribose Transferases/metabolism , Animals , Cell Differentiation , Cell Line , Chromatin/chemistry , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Methylation , Mice , Mutation , Pluripotent Stem Cells/cytology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Transcription Factors/metabolism , Trophoblasts/cytology , Trophoblasts/enzymology
4.
Development ; 138(8): 1541-50, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21389056

ABSTRACT

In XX female mammals a single X chromosome is inactivated early in embryonic development, a process that is required to equalise X-linked gene dosage relative to XY males. X inactivation is regulated by a cis-acting master switch, the Xist locus, the product of which is a large non-coding RNA that coats the chromosome from which it is transcribed, triggering recruitment of chromatin modifying factors that establish and maintain gene silencing chromosome wide. Chromosome coating and Xist RNA-mediated silencing remain poorly understood, both at the level of RNA sequence determinants and interacting factors. Here, we describe analysis of a novel targeted mutation, Xist(INV), designed to test the function of a conserved region located in exon 1 of Xist RNA during X inactivation in mouse. We show that Xist(INV) is a strong hypomorphic allele that is appropriately regulated but compromised in its ability to silence X-linked loci in cis. Inheritance of Xist(INV) on the paternal X chromosome results in embryonic lethality due to failure of imprinted X inactivation in extra-embryonic lineages. Female embryos inheriting Xist(INV) on the maternal X chromosome undergo extreme secondary non-random X inactivation, eliminating the majority of cells that express the Xist(INV) allele. Analysis of cells that express Xist(INV) RNA demonstrates reduced association of the mutant RNA to the X chromosome, suggesting that conserved sequences in the inverted region are important for Xist RNA localisation.


Subject(s)
Exons/genetics , Genes, X-Linked/genetics , RNA, Untranslated/genetics , X Chromosome Inactivation/genetics , Animals , Blotting, Northern , Cells, Cultured , Female , Fibroblasts/metabolism , Fluorescent Antibody Technique , In Situ Hybridization, Fluorescence , Male , Mice , RNA, Long Noncoding , Reverse Transcriptase Polymerase Chain Reaction
5.
Development ; 138(8): 1521-30, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21427142

ABSTRACT

During development, the growth of the embryo must be coupled to its patterning to ensure correct and timely morphogenesis. In the mouse embryo, migration of the anterior visceral endoderm (AVE) to the prospective anterior establishes the anterior-posterior (A-P) axis. By analysing the distribution of cells in S phase, M phase and G2 from the time just prior to the migration of the AVE until 18 hours after its movement, we show that there is no evidence for differential proliferation along the A-P axis of the mouse embryo. Rather, we have identified that as AVE movements are being initiated, the epiblast proliferates at a much higher rate than the visceral endoderm. We show that these high levels of proliferation in the epiblast are dependent on Nodal signalling and are required for A-P establishment, as blocking cell division in the epiblast inhibits AVE migration. Interestingly, inhibition of migration by blocking proliferation can be rescued by Dkk1. This suggests that the high levels of epiblast proliferation function to move the prospective AVE away from signals that are inhibitory to its migration. The finding that initiation of AVE movements requires a certain level of proliferation in the epiblast provides a mechanism whereby A-P axis development is coordinated with embryonic growth.


Subject(s)
Embryo, Mammalian/cytology , Endoderm/cytology , Viscera/embryology , Animals , Cell Cycle/physiology , Cell Movement/physiology , Cell Proliferation , Embryo, Mammalian/metabolism , Endoderm/metabolism , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Mice
6.
Stem Cells ; 30(12): 2732-45, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23034951

ABSTRACT

Embryonic (ES) and epiblast (EpiSC) stem cells are pluripotent but committed to an embryonic lineage fate. Conversely, trophoblast (TS) and extraembryonic endoderm (XEN) stem cells contribute predominantly to tissues of the placenta and yolk sac, respectively. Here we show that each of these four stem cell types is defined by a unique DNA methylation profile. Despite their distinct developmental origin, TS and XEN cells share key epigenomic hallmarks, chiefly characterized by robust DNA methylation of embryo-specific developmental regulators, as well as a subordinate role of 5-hydroxymethylation. We also observe a substantial methylation reinforcement of pre-existing epigenetic repressive marks that specifically occurs in extraembryonic stem cells compared to in vivo tissue, presumably due to continued high Dnmt3b expression levels. These differences establish a major epigenetic barrier between the embryonic and extraembryonic stem cell types. In addition, epigenetic lineage boundaries also separate the two extraembryonic stem cell types by mutual repression of key lineage-specific transcription factors. Thus, global DNA methylation patterns are a defining feature of each stem cell type that underpin lineage commitment and differentiative potency of early embryo-derived stem cells. Our detailed methylation profiles identify a cohort of developmentally regulated sequence elements, such as orphan CpG islands, that will be most valuable to uncover novel transcriptional regulators and pivotal "gatekeeper" genes in pluripotency and lineage differentiation.


Subject(s)
DNA Methylation , Embryonic Stem Cells/classification , Pluripotent Stem Cells/classification , Animals , Cell Differentiation/physiology , Cell Lineage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Endoderm/cytology , Female , Genomic Imprinting , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Pregnancy , Trophoblasts/cytology , X Chromosome Inactivation
7.
J Pathol ; 228(4): 554-64, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22733590

ABSTRACT

We recently reported the first evidence of placental endoplasmic reticulum (ER) stress in the pathophysiology of human intrauterine growth restriction. Here, we used a mouse model to investigate potential underlying mechanisms. Eif2s1(tm1RjK) mice, in which Ser51 of eukaryotic initiation factor 2 subunit alpha (eIF2α) is mutated, display a 30% increase in basal translation. In Eif2s1(tm1RjK) placentas, we observed increased ER stress and anomalous accumulation of glycoproteins in the endocrine junctional zone (Jz), but not in the labyrinthine zone where physiological exchange occurs. Placental and fetal weights were reduced by 15% (97 mg to 82 mg, p < 0.001) and 20% (1009 mg to 798 mg, p < 0.001), respectively. To investigate whether ER stress affects bioactivity of secreted proteins, mouse embryonic fibroblasts (MEFs) were derived from Eif2s1(tm1RjK) mutants. These MEFs exhibited ER stress, grew 50% slower, and showed reduced Akt-mTOR signalling compared to wild-type cells. Conditioned medium (CM) derived from Eif2s1(tm1RjK) MEFs failed to maintain trophoblast stem cells in a progenitor state, but the effect could be rescued by exogenous application of FGF4 and heparin. In addition, ER stress promoted accumulation of pro-Igf2 with altered glycosylation in the CM without affecting cellular levels, indicating that the protein failed to be processed after release. Igf2 is the major growth factor for placental development; indeed, activity in the Pdk1-Akt-mTOR pathways was decreased in Eif2s1(tm1RjK) placentas, indicating loss of Igf2 signalling. Furthermore, we observed premature differentiation of trophoblast progenitors at E9.5 in mutant placentas, consistent with the in vitro results and with the disproportionate development of the labyrinth and Jz seen in placentas at E18.5. Similar disproportion has been reported in the Igf2-null mouse. These results demonstrate that ER stress adversely affects placental development, and that modulation of post-translational processing, and hence bioactivity, of secreted growth factors contributes to this effect. Placental dysmorphogenesis potentially affects fetal growth through reduced exchange capacity.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Placenta/pathology , Placentation , Animals , Cell Differentiation/physiology , Disease Models, Animal , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Female , Fetal Growth Retardation/physiopathology , Glycoproteins/metabolism , Glycosylation , Humans , Mice, Inbred C57BL , Mice, Mutant Strains , Organ Size , Placenta/metabolism , Pregnancy , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism
8.
Front Cell Dev Biol ; 11: 1209928, 2023.
Article in English | MEDLINE | ID: mdl-37440923

ABSTRACT

One-carbon metabolism, including the folate cycle, has a crucial role in fetal development though its molecular function is complex and unclear. The hypomorphic Mtrr gt allele is known to disrupt one-carbon metabolism, and thus methyl group availability, leading to several developmental phenotypes (e.g., neural tube closure defects, fetal growth anomalies). Remarkably, previous studies showed that some of the phenotypes were transgenerationally inherited. Here, we explored the genome-wide epigenetic impact of one-carbon metabolism in placentas associated with fetal growth phenotypes and determined whether specific DNA methylation changes were inherited. Firstly, methylome analysis of Mtrr gt/gt homozygous placentas revealed genome-wide epigenetic instability. Several differentially methylated regions (DMRs) were identified including at the Cxcl1 gene promoter and at the En2 gene locus, which may have phenotypic implications. Importantly, we discovered hypomethylation and ectopic expression of a subset of ERV elements throughout the genome of Mtrr gt/gt placentas with broad implications for genomic stability. Next, we determined that known spermatozoan DMRs in Mtrr gt/gt males were reprogrammed in the placenta with little evidence of direct or transgenerational germline DMR inheritance. However, some spermatozoan DMRs were associated with placental gene misexpression despite normalisation of DNA methylation, suggesting the inheritance of an alternative epigenetic mechanism. Integration of published wildtype histone ChIP-seq datasets with Mtrr gt/gt spermatozoan methylome and placental transcriptome datasets point towards H3K4me3 deposition at key loci. These data suggest that histone modifications might play a role in epigenetic inheritance in this context. Overall, this study sheds light on the mechanistic complexities of one-carbon metabolism in development and epigenetic inheritance.

9.
Reprod Biomed Online ; 22(6): 529-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21498123

ABSTRACT

DNA methylation is involved in a number of important processes such as maintaining genome stability, silencing of retrotransposons, co-ordinating mono-alleleic expression of parentally imprinted genes and ensuring transcriptional repression of genes on the inactive X chromosome. Further, correct DNA methylation patterns are necessary for normal development and lineage commitment. DNA methylation provides a stable and heritable epigenetic mark. However, it can be removed, either actively or passively, during periods of reprogramming in primordial germ cells and preimplantation embryos. By combining immunofluorescence data with recent insights from genome-wide studies utilizing techniques such as Bisulphite-seq and MeDIP-ChIP, a clearer picture of the dynamic patterns of DNA methylation throughout gametogenesis, preimplantation development and early lineage commitment is beginning to emerge. The continuing use of these next-generation technologies to elucidate genome-wide methylation patterns in a variety of cellular contexts will further understanding of how this epigenetic mark contributes to lineage commitment, differentiation and pluripotency and, ultimately, to human health and disease.


Subject(s)
Blastocyst/physiology , DNA Methylation , Embryonic Development/physiology , Animals , Cell Lineage , Gametogenesis/physiology , Genomic Imprinting , Germ Cells/physiology , Humans
10.
Stem Cell Reports ; 15(6): 1301-1316, 2020 12 08.
Article in English | MEDLINE | ID: mdl-32442533

ABSTRACT

The ten-eleven translocation factor TET1 and its conferred epigenetic modification 5-hydroxymethylcytosine (5hmC) have important roles in maintaining the pluripotent state of embryonic stem cells (ESCs). We previously showed that TET1 is also essential to maintain the stem cell state of trophoblast stem cells (TSCs). Here, we establish an integrated panel of absolute 5hmC levels, genome-wide DNA methylation and hydroxymethylation patterns, transcriptomes, and TET1 chromatin occupancy in TSCs and differentiated trophoblast cells. We show that the combined presence of 5-methylcytosine (5mC) and 5hmC correlates with transcriptional activity of associated genes. Hypoxia can slow down the global loss of 5hmC that occurs upon differentiation of TSCs. Notably, unlike in ESCs and epiblast cells, most TET1-bound regions overlap with active chromatin marks and TFAP2C binding sites and demarcate putative trophoblast enhancer regions. These chromatin modification and occupancy patterns are highly informative to identify novel candidate regulators of the TSC state.


Subject(s)
DNA Methylation , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Stem Cells/metabolism , Trophoblasts/metabolism , Animals , Cell Hypoxia , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Mice , Proto-Oncogene Proteins/genetics , Stem Cells/cytology , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Trophoblasts/cytology
11.
Nat Commun ; 9(1): 4189, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30305613

ABSTRACT

The establishment of the embryonic and trophoblast lineages is a developmental decision underpinned by dramatic differences in the epigenetic landscape of the two compartments. However, it remains unknown how epigenetic information and transcription factor networks map to the 3D arrangement of the genome, which in turn may mediate transcriptional divergence between the two cell lineages. Here, we perform promoter capture Hi-C experiments in mouse trophoblast (TSC) and embryonic (ESC) stem cells to understand how chromatin conformation relates to cell-specific transcriptional programmes. We find that key TSC genes that are kept repressed in ESCs exhibit interactions between H3K27me3-marked regions in ESCs that depend on Polycomb repressive complex 1. Interactions that are prominent in TSCs are enriched for enhancer-gene contacts involving key TSC transcription factors, as well as TET1, which helps to maintain the expression of TSC-relevant genes. Our work shows that the first developmental cell fate decision results in distinct chromatin conformation patterns establishing lineage-specific contexts involving both repressive and active interactions.


Subject(s)
Cell Lineage , Chromatin/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , Animals , DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Genome , Histones/metabolism , Lysine/metabolism , Methylation , Mice , Mice, Inbred ICR , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism
12.
Stem Cell Reports ; 10(4): 1355-1368, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29576538

ABSTRACT

The ten-eleven translocation (TET) proteins are well known for their role in maintaining naive pluripotency of embryonic stem cells. Here, we demonstrate that, jointly, TET1 and TET2 also safeguard the self-renewal potential of trophoblast stem cells (TSCs) and have partially redundant roles in maintaining the epithelial integrity of TSCs. For the more abundantly expressed TET1, we show that this is achieved by binding to critical epithelial genes, notably E-cadherin, which becomes hyper-methylated and downregulated in the absence of TET1. The epithelial-to-mesenchymal transition phenotype of mutant TSCs is accompanied by centrosome duplication and separation defects. Moreover, we identify a role of TET1 in maintaining cyclin B1 stability, thereby acting as facilitator of mitotic cell-cycle progression. As a result, Tet1/2 mutant TSCs are prone to undergo endoreduplicative cell cycles leading to the formation of polyploid trophoblast giant cells. Taken together, our data reveal essential functions of TET proteins in the trophoblast lineage.


Subject(s)
Cell Cycle , Proto-Oncogene Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Trophoblasts/cytology , Animals , Cell Line , Cell Nucleus Shape , Centrosome/metabolism , Cyclin B1/metabolism , Endoreduplication , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation , Gene Knockout Techniques , Humans , Polyploidy , Protein Stability
13.
Cell Stem Cell ; 17(5): 557-68, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26412560

ABSTRACT

Trophoblast stem cells (TSCs) arise from the first cell fate decision in the developing embryo and generate extra-embryonic lineages, giving rise to the fetal portion of the placenta. Mouse embryonic and extra-embryonic lineages are strictly separated by a distinct epigenetic barrier, which is not fully overcome following expression of TSC-determining factors in embryonic stem cells. Here, we show that transient expression of Tfap2c, Gata3, Eomes, and Ets2 is sufficient to reprogram mouse embryonic fibroblasts and post-natal tail-tip-derived fibroblasts into induced TSCs (iTSCs) and surmount the epigenetic barrier separating somatic from extra-embryonic lineages. iTSCs share nearly identical morphological characteristics, gene expression profiles, and DNA methylation patterns with blastocyst-derived TSCs. Furthermore, iTSCs display transgene-independent self-renewal, differentiate along extra-embryonic lineages, and chimerize host placentas following blastocyst injection. These findings provide insights into the transcription factor networks governing TSC identity and opportunities for studying the epigenetic barriers underlying embryonic and extra-embryonic lineage segregation.


Subject(s)
Cell Transdifferentiation , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Trophoblasts/cytology , Animals , Cell Cycle , Cell Lineage , Cells, Cultured , Mice
14.
Nat Commun ; 5: 5538, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25423963

ABSTRACT

Embryonic (ES) and trophoblast (TS) stem cells reflect the first, irrevocable cell fate decision in development that is reinforced by distinct epigenetic lineage barriers. Nonetheless, ES cells can seemingly acquire TS-like characteristics upon manipulation of lineage-determining transcription factors or activation of the extracellular signal-regulated kinase 1/2 (Erk1/2) pathway. Here we have interrogated the progression of reprogramming in ES cell models with regulatable Oct4 and Cdx2 transgenes or conditional Erk1/2 activation. Although trans-differentiation into TS-like cells is initiated, lineage conversion remains incomplete in all models, underpinned by the failure to demethylate a small group of TS cell genes. Forced expression of these non-reprogrammed genes improves trans-differentiation efficiency, but still fails to confer a stable TS cell phenotype. Thus, even ES cells in ground-state pluripotency cannot fully overcome the boundaries that separate the first cell lineages but retain an epigenetic memory of their ES cell origin.


Subject(s)
Cellular Reprogramming , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Trophoblasts/cytology , Animals , CDX2 Transcription Factor , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Trophoblasts/metabolism
15.
Epigenetics Chromatin ; 4(1): 17, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21982142

ABSTRACT

BACKGROUND: Expression of Xist, the master regulator of X chromosome inactivation, is extinguished in pluripotent cells, a process that has been linked to programmed X chromosome reactivation. The key pluripotency transcription factors Nanog, Oct4 and Sox2 are implicated in Xist gene extinction, at least in part through binding to an element located in Xist intron 1. Other pathways, notably repression by the antisense RNA Tsix, may also be involved. RESULTS: Here we employ a transgene strategy to test the role of the intron 1 element and Tsix in repressing Xist in ES cells. We find that deletion of the intron 1 element causes a small increase in Xist expression and that simultaneous deletion of the antisense regulator Tsix enhances this effect. CONCLUSION: We conclude that Tsix and pluripotency factors act synergistically to repress Xist in undifferentiated embryonic stem cells. Double mutants do not exhibit maximal levels of Xist expression, indicating that other pathways also play a role.

16.
Cell Stem Cell ; 9(2): 144-55, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21816365

ABSTRACT

BMP is thought to induce hESC differentiation toward multiple lineages including mesoderm and trophoblast. The BMP-induced trophoblast phenotype is a long-standing paradox in stem cell biology. Here we readdressed BMP function in hESCs and mouse epiblast-derived cells. We found that BMP4 cooperates with FGF2 (via ERK) to induce mesoderm and to inhibit endoderm differentiation. These conditions induced cells with high levels of BRACHYURY (BRA) that coexpressed CDX2. BRA was necessary for and preceded CDX2 expression; both genes were essential for expression not only of mesodermal genes but also of trophoblast-associated genes. Maximal expression of the latter was seen in the absence of FGF but these cells coexpressed mesodermal genes and moreover they differed in cell surface and epigenetic properties from placental trophoblast. We conclude that BMP induces human and mouse pluripotent stem cells primarily to form mesoderm, rather than trophoblast, acting through BRA and CDX2.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Lineage/drug effects , Embryonic Stem Cells/cytology , Fetal Proteins/metabolism , Homeodomain Proteins/metabolism , Pluripotent Stem Cells/cytology , T-Box Domain Proteins/metabolism , Animals , CDX2 Transcription Factor , Chromones/pharmacology , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fetal Proteins/genetics , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Regulatory Networks/genetics , Glycoprotein Hormones, alpha Subunit/genetics , Glycoprotein Hormones, alpha Subunit/metabolism , Homeodomain Proteins/genetics , Humans , Keratin-7/genetics , Keratin-7/metabolism , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Morpholines/pharmacology , Neuropeptides/genetics , Neuropeptides/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Signal Transduction/drug effects , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/metabolism
17.
Curr Opin Genet Dev ; 19(2): 122-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19345091

ABSTRACT

The large non-coding RNA Xist is the master regulator of X inactivation. Xist is negatively regulated by its antisense transcript Tsix. This repressive antisense transcription across Xist operates at least in part through the modification of the chromatin environment of the locus. However Tsix is not sufficient to repress Xist in pluripotent cells and there is emerging evidence that transcription factors associated with pluripotency are involved in Tsix-independent repression. This review focuses on recent advances in this area and discusses the implications for our understanding of Xist gene regulation at the onset of X inactivation.


Subject(s)
Gene Expression Regulation, Developmental , RNA, Untranslated/genetics , X Chromosome Inactivation/genetics , Animals , Chromatin/genetics , Chromatin/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Models, Biological , Polycomb Repressive Complex 2 , RNA, Long Noncoding , Repressor Proteins/metabolism
18.
Epigenetics Chromatin ; 1(1): 2, 2008 Oct 27.
Article in English | MEDLINE | ID: mdl-19014663

ABSTRACT

BACKGROUND: X chromosome inactivation is the mechanism used in mammals to achieve dosage compensation of X-linked genes in XX females relative to XY males. Chromosome silencing is triggered in cis by expression of the non-coding RNA Xist. As such, correct regulation of the Xist gene promoter is required to establish appropriate X chromosome activity both in males and females. Studies to date have demonstrated co-transcription of an antisense RNA Tsix and low-level sense transcription prior to onset of X inactivation. The balance of sense and antisense RNA is important in determining the probability that a given Xist allele will be expressed, termed the X inactivation choice, when X inactivation commences. RESULTS: Here we investigate further the mechanism of Xist promoter regulation. We demonstrate that both sense and antisense transcription modulate Xist promoter DNA methylation in undifferentiated embryonic stem (ES) cells, suggesting a possible mechanistic basis for influencing X chromosome choice. Given the involvement of sense and antisense RNAs in promoter methylation, we investigate a possible role for the RNA interference (RNAi) pathway. We show that the Xist promoter is hypomethylated in ES cells deficient for the essential RNAi enzyme Dicer, but that this effect is probably a secondary consequence of reduced levels of de novo DNA methyltransferases in these cells. Consistent with this we find that Dicer-deficient XY and XX embryos show appropriate Xist expression patterns, indicating that Xist gene regulation has not been perturbed. CONCLUSION: We conclude that Xist promoter methylation prior to the onset of random X chromosome inactivation is influenced by relative levels of sense and antisense transcription but that this probably occurs independent of the RNAi pathway. We discuss the implications for this data in terms of understanding Xist gene regulation and X chromosome choice in random X chromosome inactivation.

SELECTION OF CITATIONS
SEARCH DETAIL