Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Nat Genet ; 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724747

ABSTRACT

Chromatin modifications are linked with regulating patterns of gene expression, but their causal role and context-dependent impact on transcription remains unresolved. Here we develop a modular epigenome editing platform that programs nine key chromatin modifications, or combinations thereof, to precise loci in living cells. We couple this with single-cell readouts to systematically quantitate the magnitude and heterogeneity of transcriptional responses elicited by each specific chromatin modification. Among these, we show that installing histone H3 lysine 4 trimethylation (H3K4me3) at promoters can causally instruct transcription by hierarchically remodeling the chromatin landscape. We further dissect how DNA sequence motifs influence the transcriptional impact of chromatin marks, identifying switch-like and attenuative effects within distinct cis contexts. Finally, we examine the interplay of combinatorial modifications, revealing that co-targeted H3K27 trimethylation (H3K27me3) and H2AK119 monoubiquitination (H2AK119ub) maximizes silencing penetrance across single cells. Our precision-perturbation strategy unveils the causal principles of how chromatin modification(s) influence transcription and dissects how quantitative responses are calibrated by contextual interactions.

2.
Nature ; 629(8012): 652-659, 2024 May.
Article in English | MEDLINE | ID: mdl-38693261

ABSTRACT

The gut microbiota operates at the interface of host-environment interactions to influence human homoeostasis and metabolic networks1-4. Environmental factors that unbalance gut microbial ecosystems can therefore shape physiological and disease-associated responses across somatic tissues5-9. However, the systemic impact of the gut microbiome on the germline-and consequently on the F1 offspring it gives rise to-is unexplored10. Here we show that the gut microbiota act as a key interface between paternal preconception environment and intergenerational health in mice. Perturbations to the gut microbiota of prospective fathers increase the probability of their offspring presenting with low birth weight, severe growth restriction and premature mortality. Transmission of disease risk occurs via the germline and is provoked by pervasive gut microbiome perturbations, including non-absorbable antibiotics or osmotic laxatives, but is rescued by restoring the paternal microbiota before conception. This effect is linked with a dynamic response to induced dysbiosis in the male reproductive system, including impaired leptin signalling, altered testicular metabolite profiles and remapped small RNA payloads in sperm. As a result, dysbiotic fathers trigger an elevated risk of in utero placental insufficiency, revealing a placental origin of mammalian intergenerational effects. Our study defines a regulatory 'gut-germline axis' in males, which is sensitive to environmental exposures and programmes offspring fitness through impacting placenta function.


Subject(s)
Disease Susceptibility , Dysbiosis , Fathers , Gastrointestinal Microbiome , Placental Insufficiency , Prenatal Injuries , Spermatozoa , Animals , Female , Male , Mice , Pregnancy , Dysbiosis/complications , Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Leptin/metabolism , Mice, Inbred C57BL , Placenta/metabolism , Placenta/physiopathology , Placental Insufficiency/etiology , Placental Insufficiency/metabolism , Placental Insufficiency/physiopathology , Pregnancy Outcome , Prenatal Injuries/etiology , Prenatal Injuries/metabolism , Prenatal Injuries/physiopathology , Signal Transduction , Spermatozoa/metabolism , Testis/metabolism , Testis/physiopathology , Disease Susceptibility/etiology
3.
Nat Cell Biol ; 25(5): 643-657, 2023 05.
Article in English | MEDLINE | ID: mdl-37106060

ABSTRACT

During embryonic development, naive pluripotent epiblast cells transit to a formative state. The formative epiblast cells form a polarized epithelium, exhibit distinct transcriptional and epigenetic profiles and acquire competence to differentiate into all somatic and germline lineages. However, we have limited understanding of how the transition to a formative state is molecularly controlled. Here we used murine embryonic stem cell models to show that ESRRB is both required and sufficient to activate formative genes. Genetic inactivation of Esrrb leads to illegitimate expression of mesendoderm and extra-embryonic markers, impaired formative expression and failure to self-organize in 3D. Functionally, this results in impaired ability to generate formative stem cells and primordial germ cells in the absence of Esrrb. Computational modelling and genomic analyses revealed that ESRRB occupies key formative genes in naive cells and throughout the formative state. In so doing, ESRRB kickstarts the formative transition, leading to timely and unbiased capacity for multi-lineage differentiation.


Subject(s)
Embryonic Stem Cells , Pluripotent Stem Cells , Mice , Animals , Cell Differentiation/genetics , Pluripotent Stem Cells/metabolism , Germ Layers/metabolism , Germ Cells/metabolism , Receptors, Estrogen/metabolism
4.
EMBO J ; 41(7): e108677, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35199868

ABSTRACT

Environmental factors can trigger cellular responses that propagate across mitosis or even generations. Perturbations to the epigenome could underpin such acquired changes, however, the extent and contexts in which modified chromatin states confer heritable memory in mammals is unclear. Here, we exploit a precision epigenetic editing strategy and forced Xist activity to programme de novo heterochromatin domains (epialleles) at endogenous loci and track their inheritance in a developmental model. We find that naïve pluripotent phases systematically erase ectopic domains of heterochromatin via active mechanisms, which likely acts as an intergenerational safeguard against transmission of epialleles. Upon lineage specification, however, acquired chromatin states can be probabilistically inherited under selectively favourable conditions, including propagation of p53 silencing through in vivo development. Using genome-wide CRISPR screening, we identify molecular factors that restrict heritable memory of epialleles in naïve pluripotent cells, and demonstrate that removal of chromatin factor Dppa2 unlocks the potential for epigenetic inheritance uncoupled from DNA sequence. Our study outlines a mechanistic basis for how epigenetic inheritance is constrained in mammals, and reveals genomic and developmental contexts in which heritable memory is feasible.


Subject(s)
Epigenesis, Genetic , Epigenomics , Animals , Chromatin , Genome , Heterochromatin , Mammals/genetics
5.
Bioessays ; 43(5): e2000316, 2021 05.
Article in English | MEDLINE | ID: mdl-33724509

ABSTRACT

How epigenetic mechanisms regulate genome output and response to stimuli is a fundamental question in development and disease. Past decades have made tremendous progress in deciphering the regulatory relationships involved by correlating aggregated (epi)genomics profiles with global perturbations. However, the recent development of epigenetic editing technologies now enables researchers to move beyond inferred conclusions, towards explicit causal reasoning, through 'programing' precise chromatin perturbations in single cells. Here, we first discuss the major unresolved questions in the epigenetics field that can be addressed by programable epigenome editing, including the context-dependent function and memory of chromatin states. We then describe the epigenetic editing toolkit focusing on CRISPR-based technologies, and highlight its achievements, drawbacks and promise. Finally, we consider the potential future application of epigenetic editing to the study and treatment of specific disease conditions.


Subject(s)
CRISPR-Cas Systems , Gene Editing , CRISPR-Cas Systems/genetics , Chromatin/genetics , Epigenesis, Genetic/genetics , Epigenomics
6.
Methods Mol Biol ; 2214: 91-108, 2021.
Article in English | MEDLINE | ID: mdl-32944905

ABSTRACT

Knockout CRISPR screening enables the unbiased discovery of genes with a functional role in almost any cellular or molecular process of interest. The approach couples a genome-scale library of guide RNA (gRNA), the Cas9 endonuclease, and a faithful phenotypic read-out to systematically identify candidate genes via their loss-of-function effect. Here we provide a detailed description of the CRISPR screen protocol and outline how to apply it to decipher the gene networks that underlie developmental cell fate decisions. As a paradigm we use the in vitro model of cell state transition(s) from naive pluripotency to primordial germ cell (PGC) fate, exploiting the Stella-GFP:Esg1-tdTomato (SGET) mouse ESC line. The principles in this protocol can be readily adapted to characterize lineage regulators for other cell fate models and/or for other species.


Subject(s)
CRISPR-Cas Systems , Embryonic Germ Cells/cytology , Mouse Embryonic Stem Cells/cytology , Animals , Cell Differentiation , Cell Line , Embryonic Germ Cells/metabolism , Gene Regulatory Networks , HEK293 Cells , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , RNA, Guide, Kinetoplastida/genetics , Transduction, Genetic
7.
Cell Stem Cell ; 28(2): 209-216.e4, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33207217

ABSTRACT

Cell differentiation typically occurs with concomitant shape transitions to enable specialized functions. To adopt a different shape, cells need to change the mechanical properties of their surface. However, whether cell surface mechanics control the process of differentiation has been relatively unexplored. Here we show that membrane mechanics gate exit from naive pluripotency of mouse embryonic stem cells. By measuring membrane tension during early differentiation, we find that naive stem cells release their plasma membrane from the underlying actin cortex when transitioning to a primed state. By mechanically tethering the plasma membrane to the cortex by enhancing Ezrin activity or expressing a synthetic signaling-inert linker, we demonstrate that preventing this detachment forces stem cells to retain their naive pluripotent identity. We thus identify a decrease in membrane-to-cortex attachment as a new cell-intrinsic mechanism that is essential for stem cells to exit pluripotency.


Subject(s)
Embryonic Stem Cells , Mouse Embryonic Stem Cells , Animals , Cell Differentiation , Cell Membrane , Mice , Signal Transduction
8.
Nat Struct Mol Biol ; 27(8): 706-716, 2020 08.
Article in English | MEDLINE | ID: mdl-32572256

ABSTRACT

Early mammalian development entails genome-wide epigenome remodeling, including DNA methylation erasure and reacquisition, which facilitates developmental competence. To uncover the mechanisms that orchestrate DNA methylation dynamics, we coupled a single-cell ratiometric DNA methylation reporter with unbiased CRISPR screening in murine embryonic stem cells (ESCs). We identify key genes and regulatory pathways that drive global DNA hypomethylation, and characterize roles for Cop1 and Dusp6. We also identify Dppa2 and Dppa4 as essential safeguards of focal epigenetic states. In their absence, developmental genes and evolutionarily young LINE1 elements, which are specifically bound by DPPA2, lose H3K4me3 and gain ectopic de novo DNA methylation in pluripotent cells. Consequently, lineage-associated genes and LINE1 acquire a repressive epigenetic memory, which renders them incompetent for activation during future lineage specification. Dppa2/4 thereby sculpt the pluripotent epigenome by facilitating H3K4me3 and bivalency to counteract de novo methylation, a function co-opted by evolutionarily young LINE1 to evade epigenetic decommissioning.


Subject(s)
DNA Methylation , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , CRISPR-Cas Systems , Cell Line , Epigenome , Gene Expression Regulation, Developmental , Long Interspersed Nucleotide Elements , Mice , Mouse Embryonic Stem Cells/cytology , Nuclear Proteins/metabolism , Single-Cell Analysis , Transcription Factors/metabolism
9.
Sci Rep ; 9(1): 19214, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31844114

ABSTRACT

Gene delivery using vector or viral-based methods is often limited by technical and safety barriers. A promising alternative that circumvents these shortcomings is the direct delivery of proteins into cells. Here we introduce a non-viral, ligand-mediated protein delivery system capable of selectively targeting primary skin cells in-vivo. Using orthologous self-labelling tags and chemical cross-linkers, we conjugate large proteins to ligands that bind their natural receptors on the surface of keratinocytes. Targeted CRE-mediated recombination was achieved by delivery of ligand cross-linked CRE protein to the skin of transgenic reporter mice, but was absent in mice lacking the ligand's cell surface receptor. We further show that ligands mediate the intracellular delivery of Cas9 allowing for CRISPR-mediated gene editing in the skin more efficiently than adeno-associated viral gene delivery. Thus, a ligand-based system enables the effective and receptor-specific delivery of large proteins and may be applied to the treatment of skin-related genetic diseases.


Subject(s)
Proteins/genetics , Proteins/metabolism , Animals , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Dependovirus/genetics , Gene Editing/methods , Gene Transfer Techniques , Genetic Therapy/methods , Keratinocytes/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Skin/metabolism
10.
Nat Commun ; 9(1): 5328, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30538240

ABSTRACT

Ufuk Günesdogan was incorrectly associated with Center for Genetic Analysis of Behaviour, National Institute for Physiological Sciences, 5-1 Higashiyama Myodaiji, Okazaki, Aichi 444-8787, Japan and Toshihiro Kobayashi was incorrectly associated with Department of Developmental Biology, University of Göttingen, Göttingen Center for Molecular Biosciences, Justus-von-Liebig Weg 11, 37077 Göttingen, Germany. This has now been corrected in both the PDF and HTML versions of the Article.

11.
Nat Commun ; 9(1): 4292, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30327475

ABSTRACT

Early mammalian development entails transit through naive pluripotency towards post-implantation epiblast, which subsequently gives rise to primordial germ cells (PGC), the founding germline population. To investigate these cell fate transitions, we developed a compound-reporter to track cellular identity in a model of PGC specification (PGC-like cells; PGCLC), and coupled it with genome-wide CRISPR screening. We identify key genes both for exit from pluripotency and for acquisition of PGC fate, and characterise a central role for the transcription regulators Nr5a2 and Zfp296 in germline ontogeny. Abrogation of these genes results in widespread activation (Nr5a2-/-) or inhibition (Zfp296-/-) of WNT pathway factors in PGCLC. This leads to aberrant upregulation of the somatic programme or failure to activate germline genes, respectively, and consequently loss of germ cell identity. Our study places Zfp296 and Nr5a2 as key components of an expanded PGC gene regulatory network, and outlines a transferable strategy for identifying critical regulators of complex cell fate decisions.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Expression Regulation, Developmental , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins/genetics , Embryonic Development/genetics , Genes, Reporter , Green Fluorescent Proteins/genetics , Mice , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Wnt Proteins/genetics
12.
Stem Cell Reports ; 8(6): 1645-1658, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28591649

ABSTRACT

Embryonic stem cells (ESCs) are characterized by the pluripotent capacity to generate all embryonic lineages. Here, we show that ESCs can occupy a spectrum of distinct transcriptional and epigenetic states in response to varied extrinsic conditions. This spectrum broadly corresponds to a developmental continuum of pluripotency and is coupled with a gradient of increasing global DNA methylation. Each pluripotent state is linked with activation of distinct classes of transposable elements (TEs), which in turn influence ESCs through generating chimeric transcripts. Moreover, varied ESC culture parameters differentially license heterogeneous activation of master lineage regulators, including Sox1, Gata4, or Blimp1, and influence differentiation. Activation of Blimp1 is prevalent in 2i (without LIF) conditions, and marks a dynamic primordial germ cell (PGC)-like sub-state that is directly repressed by Klf4 downstream of LIF/STAT3 signaling. Thus, extrinsic cues establish a spectrum of pluripotent states, in part by modulating sub-populations, as well as directing the transcriptome, epigenome, and TE.


Subject(s)
DNA Transposable Elements/genetics , Pluripotent Stem Cells/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Line , Cell Lineage , DNA Methylation , GATA4 Transcription Factor/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukemia Inhibitory Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Positive Regulatory Domain I-Binding Factor 1/metabolism , Principal Component Analysis , SOXB1 Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
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
14.
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
15.
Cell ; 161(6): 1453-67, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046444

ABSTRACT

Resetting of the epigenome in human primordial germ cells (hPGCs) is critical for development. We show that the transcriptional program of hPGCs is distinct from that in mice, with co-expression of somatic specifiers and naive pluripotency genes TFCP2L1 and KLF4. This unique gene regulatory network, established by SOX17 and BLIMP1, drives comprehensive germline DNA demethylation by repressing DNA methylation pathways and activating TET-mediated hydroxymethylation. Base-resolution methylome analysis reveals progressive DNA demethylation to basal levels in week 5-7 in vivo hPGCs. Concurrently, hPGCs undergo chromatin reorganization, X reactivation, and imprint erasure. Despite global hypomethylation, evolutionarily young and potentially hazardous retroelements, like SVA, remain methylated. Remarkably, some loci associated with metabolic and neurological disorders are also resistant to DNA demethylation, revealing potential for transgenerational epigenetic inheritance that may have phenotypic consequences. We provide comprehensive insight on early human germline transcriptional network and epigenetic reprogramming that subsequently impacts human development and disease.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Genome, Human , Germ Cells/metabolism , Animals , DNA Methylation , Embryo, Mammalian/metabolism , Female , Humans , Kruppel-Like Factor 4 , Male , Mice , Promoter Regions, Genetic , Retroelements
16.
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
17.
Cell Stem Cell ; 15(4): 416-430, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25280218

ABSTRACT

Pluripotency is the remarkable capacity of a single cell to engender all the specialized cell types of an adult organism. This property can be captured indefinitely through derivation of self-renewing embryonic stem cells (ESCs), which represent an invaluable platform to investigate cell fate decisions and disease. Recent advances have revealed that manipulation of distinct signaling cues can render ESCs in a uniform "ground state" of pluripotency, which more closely recapitulates the pluripotent naive epiblast. Here we discuss the extrinsic and intrinsic regulatory principles that underpin the nature of pluripotency and consider the emerging spectrum of pluripotent states.


Subject(s)
Gene Expression Regulation , Pluripotent Stem Cells/metabolism , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Humans , Models, Biological , Pluripotent Stem Cells/cytology , Signal Transduction/genetics
18.
Mol Cell ; 55(2): 319-31, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25038413

ABSTRACT

Cell populations can be strikingly heterogeneous, composed of multiple cellular states, each exhibiting stochastic noise in its gene expression. A major challenge is to disentangle these two types of variability and to understand the dynamic processes and mechanisms that control them. Embryonic stem cells (ESCs) provide an ideal model system to address this issue because they exhibit heterogeneous and dynamic expression of functionally important regulatory factors. We analyzed gene expression in individual ESCs using single-molecule RNA-FISH and quantitative time-lapse movies. These data discriminated stochastic switching between two coherent (correlated) gene expression states and burst-like transcriptional noise. We further showed that the "2i" signaling pathway inhibitors modulate both types of variation. Finally, we found that DNA methylation plays a key role in maintaining these metastable states. Together, these results show how ESC gene expression states and dynamics arise from a combination of intrinsic noise, coherent cellular states, and epigenetic regulation.


Subject(s)
DNA Methylation , Embryonic Stem Cells/metabolism , Transcriptome , Animals , Cells, Cultured , Epigenesis, Genetic , Gene Expression Profiling , In Situ Hybridization, Fluorescence , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Single-Cell Analysis , Time-Lapse Imaging
19.
Stem Cell Reports ; 1(6): 518-31, 2013.
Article in English | MEDLINE | ID: mdl-24371807

ABSTRACT

Pluripotent stem cells (PSCs) occupy a spectrum of reversible molecular states ranging from a naive ground-state in 2i, to metastable embryonic stem cells (ESCs) in serum, to lineage-primed epiblast stem cells (EpiSCs). To investigate the role of DNA methylation (5mC) across distinct pluripotent states, we mapped genome-wide 5mC and 5-hydroxymethycytosine (5hmC) in multiple PSCs. Ground-state ESCs exhibit an altered distribution of 5mC and 5hmC at regulatory elements and dramatically lower absolute levels relative to ESCs in serum. By contrast, EpiSCs exhibit increased promoter 5mC coupled with reduced 5hmC, which contributes to their developmental restriction. Switch to 2i triggers rapid onset of both the ground-state gene expression program and global DNA demethylation. Mechanistically, repression of de novo methylases by PRDM14 drives DNA demethylation at slow kinetics, whereas TET1/TET2-mediated 5hmC conversion enhances both the rate and extent of hypomethylation. These processes thus act synergistically during transition to ground-state pluripotency to promote a robust hypomethylated state.


Subject(s)
Cell Differentiation/genetics , DNA Methylation , Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , DNA-Binding Proteins/genetics , Dioxygenases , Embryonic Stem Cells , Female , Gene Knockout Techniques , Genomic Imprinting , Male , Mice , Proto-Oncogene Proteins/genetics
20.
EMBO Rep ; 14(7): 629-37, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23670199

ABSTRACT

Primordial germ cells (PGCs) and somatic cells originate from postimplantation epiblast cells in mice. As pluripotency is lost upon differentiation of somatic lineages, a naive epigenome and the pluripotency network are re-established during PGC development. Here we demonstrate that Prdm14 contributes not only to PGC specification, but also to naive pluripotency in embryonic stem (ES) cells by repressing the DNA methylation machinery and fibroblast growth factor (FGF) signalling. This indicates a critical role for Prdm14 in programming PGCs and promoting pluripotency in ES cells.


Subject(s)
Embryonic Stem Cells/metabolism , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Animals , Cell Differentiation , DNA Methylation , DNA-Binding Proteins , Embryonic Stem Cells/cytology , Fibroblast Growth Factors/metabolism , Gene Expression Profiling , Germ Cells/cytology , Germ Layers/cytology , Germ Layers/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Pluripotent Stem Cells/cytology , RNA-Binding Proteins , Signal Transduction , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...