Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Cell ; 149(3): 590-604, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22541430

ABSTRACT

Mouse embryonic stem (ES) cells grown in serum exhibit greater heterogeneity in morphology and expression of pluripotency factors than ES cells cultured in defined medium with inhibitors of two kinases (Mek and GSK3), a condition known as "2i" postulated to establish a naive ground state. We show that the transcriptome and epigenome profiles of serum- and 2i-grown ES cells are distinct. 2i-treated cells exhibit lower expression of lineage-affiliated genes, reduced prevalence at promoters of the repressive histone modification H3K27me3, and fewer bivalent domains, which are thought to mark genes poised for either up- or downregulation. Nonetheless, serum- and 2i-grown ES cells have similar differentiation potential. Precocious transcription of developmental genes in 2i is restrained by RNA polymerase II promoter-proximal pausing. These findings suggest that transcriptional potentiation and a permissive chromatin context characterize the ground state and that exit from it may not require a metastable intermediate or multilineage priming.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Histone Code , Transcription, Genetic , Animals , Cell Differentiation , Epigenesis, Genetic , Genes, myc , Histones/metabolism , Methylation , Mice , RNA Polymerase II/metabolism , Transcriptome
2.
Trends Genet ; 38(1): 82-96, 2022 01.
Article in English | MEDLINE | ID: mdl-34304914

ABSTRACT

DNA methylation has long been considered the primary epigenetic mediator of genomic imprinting in mammals. Recent epigenetic profiling during early mouse development revealed the presence of domains of trimethylation of lysine 27 on histone H3 (H3K27me3) and chromatin compaction specifically at the maternally derived allele, independent of DNA methylation. Within these domains, genes are exclusively expressed from the paternally derived allele. This novel mechanism of noncanonical imprinting plays a key role in the development of mouse extraembryonic tissues and in the regulation of imprinted X-chromosome inactivation, highlighting the importance of parentally inherited epigenetic histone modifications. Here, we discuss the mechanisms underlying H3K27me3-mediated noncanonical imprinting in perspective of the dynamic chromatin landscape during early mouse development and explore evolutionary origins of noncanonical imprinting.


Subject(s)
Genomic Imprinting , Histones , Animals , Chromatin/genetics , DNA Methylation/genetics , Genomic Imprinting/genetics , Histone Code , Histones/genetics , Histones/metabolism , Mice
3.
Cell ; 142(6): 967-80, 2010 Sep 17.
Article in English | MEDLINE | ID: mdl-20850016

ABSTRACT

Trimethyl-lysine (me3) modifications on histones are the most stable epigenetic marks and they control chromatin-mediated regulation of gene expression. Here, we determine proteins that bind these marks by high-accuracy, quantitative mass spectrometry. These chromatin "readers" are assigned to complexes by interaction proteomics of full-length BAC-GFP-tagged proteins. ChIP-Seq profiling identifies their genomic binding sites, revealing functional properties. Among the main findings, the human SAGA complex binds to H3K4me3 via a double Tudor-domain in the C terminus of Sgf29, and the PWWP domain is identified as a putative H3K36me3 binding motif. The ORC complex, including LRWD1, binds to the three most prominent transcriptional repressive lysine methylation sites. Our data reveal a highly adapted interplay between chromatin marks and their associated protein complexes. Reading specific trimethyl-lysine sites by specialized complexes appears to be a widespread mechanism to mediate gene expression.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Histone Code , Gene Expression Regulation , Genome-Wide Association Study , HeLa Cells , Histone Acetyltransferases/metabolism , Humans , Lysine/metabolism , Mass Spectrometry , Methylation , Proteomics/methods
4.
Genome Res ; 31(5): 919-933, 2021 05.
Article in English | MEDLINE | ID: mdl-33707229

ABSTRACT

Epigenetic profiling by chromatin immunoprecipitation followed by sequencing (ChIP-seq) has become a powerful tool for genome-wide identification of regulatory elements, for defining transcriptional regulatory networks, and for screening for biomarkers. However, the ChIP-seq protocol for low-input samples is laborious and time-consuming and suffers from experimental variation, resulting in poor reproducibility and low throughput. Although prototypic microfluidic ChIP-seq platforms have been developed, these are poorly transferable as they require sophisticated custom-made equipment and in-depth microfluidic and ChIP expertise, while lacking parallelization. To enable standardized, automated ChIP-seq profiling of low-input samples, we constructed microfluidic PDMS-based plates capable of performing 24 sensitive ChIP reactions within 30 min of hands-on time and 4.5 h of machine-running time. These disposable plates can be conveniently loaded into a widely available controller for pneumatics and thermocycling. In light of the plug and play (PnP) ChIP plates and workflow, we named our procedure PnP-ChIP-seq. We show high-quality ChIP-seq on hundreds to a few thousand of cells for all six post-translational histone modifications that are included in the International Human Epigenome Consortium set of reference epigenomes. PnP-ChIP-seq robustly detects epigenetic differences on promoters and enhancers between naive and more primed mouse embryonic stem cells (mESCs). Furthermore, we used our platform to generate epigenetic profiles of rare subpopulations of mESCs that resemble the two-cell stage of embryonic development. PnP-ChIP-seq allows nonexpert laboratories worldwide to conveniently run robust, standardized ChIP-seq, whereas its high throughput, consistency, and sensitivity pave the way toward large-scale profiling of precious sample types such as rare subpopulations of cells or biopsies.


Subject(s)
Histones , Microfluidics , Animals , Chromatin , Chromatin Immunoprecipitation/methods , Chromatin Immunoprecipitation Sequencing , High-Throughput Nucleotide Sequencing/methods , Histones/genetics , Mice , Reproducibility of Results
5.
Exp Dermatol ; 30(8): 1023-1032, 2021 08.
Article in English | MEDLINE | ID: mdl-32681572

ABSTRACT

The epidermal compartment of the skin is regenerated constantly by proliferation of epidermal keratinocytes. Differentiation of a subset of these keratinocytes allows the epidermis to retain its barrier properties. Regulation of keratinocyte fate-whether to remain proliferative or terminally differentiate-is complex and not fully understood. The objective of our study was to assess if DNA methylation changes contribute to the regulation of keratinocyte fate. We employed genome-wide MethylationEPIC beadchip array measuring approximately 850 000 probes combined with RNA sequencing of in vitro cultured non-differentiated and terminally differentiated adult human primary keratinocytes. We did not observe a correlation between methylation status and transcriptome changes. Moreover, only two differentially methylated probes were detected, of which one was located in the TRIM29 gene. Although TRIM29 knock-down resulted in lower expression levels of terminal differentiation genes, these changes were minor. From these results, we conclude that-in our in vitro experimental setup-it is unlikely that changes in DNA methylation have an important regulatory role in terminal keratinocyte differentiation.


Subject(s)
Cell Differentiation/genetics , DNA Methylation/genetics , Epigenome/genetics , Keratinocytes/metabolism , Adult , DNA-Binding Proteins/genetics , Humans , Transcription Factors/genetics
6.
Development ; 144(7): 1221-1234, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28174249

ABSTRACT

Mouse embryonic stem (ES) cells are locked into self-renewal by shielding from inductive cues. Release from this ground state in minimal conditions offers a system for delineating developmental progression from naïve pluripotency. Here, we examine the initial transition process. The ES cell population behaves asynchronously. We therefore exploited a short-half-life Rex1::GFP reporter to isolate cells either side of exit from naïve status. Extinction of ES cell identity in single cells is acute. It occurs only after near-complete elimination of naïve pluripotency factors, but precedes appearance of lineage specification markers. Cells newly departed from the ES cell state display features of early post-implantation epiblast and are distinct from primed epiblast. They also exhibit a genome-wide increase in DNA methylation, intermediate between early and late epiblast. These findings are consistent with the proposition that naïve cells transition to a distinct formative phase of pluripotency preparatory to lineage priming.


Subject(s)
Cell Tracking , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Lineage , Cell Self Renewal , DNA Methylation/genetics , Down-Regulation , Embryo, Mammalian/cytology , Embryonic Stem Cells/metabolism , Genes, Reporter , Germ Layers/cytology , Kinetics , Mice , Pluripotent Stem Cells/metabolism , Stem Cell Transplantation , Transcription Factors/metabolism , Transcription, Genetic
7.
Proteomics ; 19(14): e1900047, 2019 07.
Article in English | MEDLINE | ID: mdl-31219242

ABSTRACT

Pluripotency can be captured in vitro in the form of Embryonic Stem Cells (ESCs). These ESCs can be either maintained in the unrestricted "naïve" state of pluripotency, adapted to developmentally more constrained "primed" pluripotency or differentiated towards each of the three germ layers. Epigenetic protein complexes and transcription factors have been shown to specify and instruct transitions from ESCs to distinct cell states. In this study, proteomic profiling of the chromatin landscape by chromatin enrichment for proteomics (ChEP) is used in mouse naive pluripotent ESCs, primed pluripotent Epiblast stem cells (EpiSCs), and cells in early stages of differentiation. A comprehensive overview of epigenetic protein complexes associated with the chromatin is provided and proteins associated with the maintenance and loss of pluripotency are identified. The data reveal major compositional alterations of epigenetic complexes during priming and differentiation of naïve pluripotent ESCs. These results contribute to the understanding of ESC differentiation and provide a framework for future studies of lineage commitment of ESCs.


Subject(s)
DNA-Binding Proteins/metabolism , Mass Spectrometry/methods , Animals , Humans , Pluripotent Stem Cells/metabolism , Proteomics/methods , Transcription Factors/metabolism
8.
Nucleic Acids Res ; 45(21): e174, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28981838

ABSTRACT

Mouse embryonic stem (ES) cells are a popular model system to study biological processes, though uncovering recessive phenotypes requires inactivating both alleles. Building upon resources from the International Knockout Mouse Consortium (IKMC), we developed a targeting vector for second allele inactivation in conditional-ready IKMC 'knockout-first' ES cell lines. We applied our technology to several epigenetic regulators, recovering bi-allelic targeted clones with a high efficiency of 60% and used Flp recombinase to restore expression in two null cell lines to demonstrate how our system confirms causality through mutant phenotype reversion. We designed our strategy to select against re-targeting the 'knockout-first' allele and identify essential genes in ES cells, including the histone methyltransferase Setdb1. For confirmation, we exploited the flexibility of our system, enabling tamoxifen inducible conditional gene ablation while controlling for genetic background and tamoxifen effects. Setdb1 ablated ES cells exhibit severe growth inhibition, which is not rescued by exogenous Nanog expression or culturing in naive pluripotency '2i' media, suggesting that the self-renewal defect is mediated through pluripotency network independent pathways. Our strategy to generate null mutant mouse ES cells is applicable to thousands of genes and repurposes existing IKMC Intermediate Vectors.


Subject(s)
Alleles , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Gene Knockout Techniques/methods , Animals , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Gene Expression , Genetic Vectors , Histone-Lysine N-Methyltransferase/genetics , Mice , Polycomb Repressive Complex 2/genetics
9.
Nat Chem Biol ; 17(1): 6-7, 2021 01.
Article in English | MEDLINE | ID: mdl-32807970
10.
Development ; 141(3): 526-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24423662

ABSTRACT

Trimethylation of histone H3 lysine 4 (H3K4me3) at the promoters of actively transcribed genes is a universal epigenetic mark and a key product of Trithorax group action. Here, we show that Mll2, one of the six Set1/Trithorax-type H3K4 methyltransferases in mammals, is required for trimethylation of bivalent promoters in mouse embryonic stem cells. Mll2 is bound to bivalent promoters but also to most active promoters, which do not require Mll2 for H3K4me3 or mRNA expression. By contrast, the Set1 complex (Set1C) subunit Cxxc1 is primarily bound to active but not bivalent promoters. This indicates that bivalent promoters rely on Mll2 for H3K4me3 whereas active promoters have more than one bound H3K4 methyltransferase, including Set1C. Removal of Mll1, sister to Mll2, had almost no effect on any promoter unless Mll2 was also removed, indicating functional backup between these enzymes. Except for a subset, loss of H3K4me3 on bivalent promoters did not prevent responsiveness to retinoic acid, thereby arguing against a priming model for bivalency. In contrast, we propose that Mll2 is the pioneer trimethyltransferase for promoter definition in the naïve epigenome and that Polycomb group action on bivalent promoters blocks the premature establishment of active, Set1C-bound, promoters.


Subject(s)
Embryonic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Promoter Regions, Genetic , Animals , Binding Sites/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Chromosomes, Artificial, Bacterial/metabolism , Embryonic Stem Cells/drug effects , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Methylation/drug effects , Mice , Models, Biological , Myeloid-Lymphoid Leukemia Protein/deficiency , Protein Binding/drug effects , Protein Binding/genetics , Transgenes/genetics , Tretinoin/pharmacology
11.
Nature ; 476(7361): 467-71, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21832993

ABSTRACT

Cohesin enables post-replicative DNA repair and chromosome segregation by holding sister chromatids together from the time of DNA replication in S phase until mitosis. There is growing evidence that cohesin also forms long-range chromosomal cis-interactions and may regulate gene expression in association with CTCF, mediator or tissue-specific transcription factors. Human cohesinopathies such as Cornelia de Lange syndrome are thought to result from impaired non-canonical cohesin functions, but a clear distinction between the cell-division-related and cell-division-independent functions of cohesion--as exemplified in Drosophila--has not been demonstrated in vertebrate systems. To address this, here we deleted the cohesin locus Rad21 in mouse thymocytes at a time in development when these cells stop cycling and rearrange their T-cell receptor (TCR) α locus (Tcra). Rad21-deficient thymocytes had a normal lifespan and retained the ability to differentiate, albeit with reduced efficiency. Loss of Rad21 led to defective chromatin architecture at the Tcra locus, where cohesion-binding sites flank the TEA promoter and the Eα enhancer, and demarcate Tcra from interspersed Tcrd elements and neighbouring housekeeping genes. Cohesin was required for long-range promoter-enhancer interactions, Tcra transcription, H3K4me3 histone modifications that recruit the recombination machinery and Tcra rearrangement. Provision of pre-rearranged TCR transgenes largely rescued thymocyte differentiation, demonstrating that among thousands of potential target genes across the genome, defective Tcra rearrangement was limiting for the differentiation of cohesin-deficient thymocytes. These findings firmly establish a cell-division-independent role for cohesin in Tcra locus rearrangement and provide a comprehensive account of the mechanisms by which cohesin enables cellular differentiation in a well-characterized mammalian system.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Differentiation , Chromosomal Proteins, Non-Histone/metabolism , Gene Rearrangement, T-Lymphocyte , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Thymus Gland/cytology , Animals , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins , Gene Expression Regulation , Gene Rearrangement, T-Lymphocyte/genetics , Genes, RAG-1/genetics , Mice , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phosphoproteins/deficiency , Phosphoproteins/genetics , Recombinases/metabolism , Thymus Gland/metabolism , Transcription, Genetic , Cohesins
12.
Biochim Biophys Acta ; 1839(3): 129-37, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24096207

ABSTRACT

The use of mouse embryonic stem cells (ESCs) has provided invaluable insights into transcription and epigenetic regulation of pluripotency and self-renewal. Many of these insights were gained in mouse ESCs that are derived and maintained using serum, either on feeder cells or supplemented with the cytokine leukemia inhibitory factor (LIF). These 'serum' ESCs are in a metastable state characterized by the expression of many lineage-specifying genes. The use of two small-molecule kinase inhibitors (2i), targeting mitogen-activated protein kinase (MEK) and glycogen synthase kinase-3 (GSK3), has enabled derivation of mouse ESCs in defined serum-free conditions. These '2i' ESCs are more homogeneous in morphology and gene expression than serum ESCs, and are postulated to represent the ground state of pluripotency. Recent studies have shown that the epigenome and transcriptome of 2i and serum ESCs are markedly different, suggesting that these ESCs represent two distinct states of pluripotency regulated by different factors and pathways. There is growing evidence that the 2i ESCs closely parallel the early blastocyst cells of the inner cell mass (ICM) or even earlier stages, while serum cells possibly reflect later stages. In this review, we will focus on the difference in chromatin structure, transcription regulation and cell cycle regulation between ground state pluripotent 2i ESCs and serum ESCs, and compare to corresponding data in embryos if available. This article is part of a Special Issue entitled: Chromatin and epigenetic regulation of animal development.


Subject(s)
Cell Cycle/physiology , Chromatin Assembly and Disassembly/physiology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/physiology , Pluripotent Stem Cells/metabolism , Transcriptome/physiology , Animals , Embryonic Stem Cells/cytology , Humans , Mice , Pluripotent Stem Cells/cytology
13.
Genome Res ; 22(6): 1128-38, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22466170

ABSTRACT

Cross-talk between DNA methylation and histone modifications drives the establishment of composite epigenetic signatures and is traditionally studied using correlative rather than direct approaches. Here, we present sequential ChIP-bisulfite-sequencing (ChIP-BS-seq) as an approach to quantitatively assess DNA methylation patterns associated with chromatin modifications or chromatin-associated factors directly. A chromatin-immunoprecipitation (ChIP)-capturing step is used to obtain a restricted representation of the genome occupied by the epigenetic feature of interest, for which a single-base resolution DNA methylation map is then generated. When applied to H3 lysine 27 trimethylation (H3K27me3), we found that H3K27me3 and DNA methylation are compatible throughout most of the genome, except for CpG islands, where these two marks are mutually exclusive. Further ChIP-BS-seq-based analysis in Dnmt triple-knockout (TKO) embryonic stem cells revealed that total loss of CpG methylation is associated with alteration of H3K27me3 levels throughout the genome: H3K27me3 in localized peaks is decreased while broad local enrichments (BLOCs) of H3K27me3 are formed. At an even broader scale, these BLOCs correspond to regions of high DNA methylation in wild-type ES cells, suggesting that DNA methylation prevents H3K27me3 deposition locally and at a megabase scale. Our strategy provides a unique way of investigating global interdependencies between DNA methylation and other chromatin features.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/genetics , CpG Islands , DNA Methylation , Histones/metabolism , Sequence Analysis, DNA/methods , Animals , Cell Line , Cell Line, Tumor , Chromatin/drug effects , Colonic Neoplasms/genetics , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Gene Knockout Techniques , Genomics/methods , Humans , Lysine/metabolism , Mice , Sulfites/pharmacology
14.
PLoS Biol ; 9(1): e1000569, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21245904

ABSTRACT

The epigenetic modification of chromatin structure and its effect on complex neuronal processes like learning and memory is an emerging field in neuroscience. However, little is known about the "writers" of the neuronal epigenome and how they lay down the basis for proper cognition. Here, we have dissected the neuronal function of the Drosophila euchromatin histone methyltransferase (EHMT), a member of a conserved protein family that methylates histone 3 at lysine 9 (H3K9). EHMT is widely expressed in the nervous system and other tissues, yet EHMT mutant flies are viable. Neurodevelopmental and behavioral analyses identified EHMT as a regulator of peripheral dendrite development, larval locomotor behavior, non-associative learning, and courtship memory. The requirement for EHMT in memory was mapped to 7B-Gal4 positive cells, which are, in adult brains, predominantly mushroom body neurons. Moreover, memory was restored by EHMT re-expression during adulthood, indicating that cognitive defects are reversible in EHMT mutants. To uncover the underlying molecular mechanisms, we generated genome-wide H3K9 dimethylation profiles by ChIP-seq. Loss of H3K9 dimethylation in EHMT mutants occurs at 5% of the euchromatic genome and is enriched at the 5' and 3' ends of distinct classes of genes that control neuronal and behavioral processes that are corrupted in EHMT mutants. Our study identifies Drosophila EHMT as a key regulator of cognition that orchestrates an epigenetic program featuring classic learning and memory genes. Our findings are relevant to the pathophysiological mechanisms underlying Kleefstra Syndrome, a severe form of intellectual disability caused by mutations in human EHMT1, and have potential therapeutic implications. Our work thus provides novel insights into the epigenetic control of cognition in health and disease.


Subject(s)
Drosophila/genetics , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Animals , Courtship , DNA/metabolism , Dendrites/metabolism , Drosophila/growth & development , Drosophila/physiology , Euchromatin/chemistry , Euchromatin/metabolism , Gene Expression Profiling , Histone-Lysine N-Methyltransferase/genetics , Humans , Larva , Learning , Locomotion , Memory , Methylation , Nervous System/growth & development , Nervous System/metabolism , Phylogeny , Sequence Deletion
15.
Stem Cell Reports ; 17(5): 1009-1011, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35545021

ABSTRACT

Epigenetic enzymes are critically involved in gene regulation during lineage commitment. In this issue of Stem Cell Reports, Zhu et al. (2022) unravel extensive redundancy between subunits of the epigenetic regulatory Polycomb Repressive Complex 1 using a systematic knockout strategy in mouse embryonic stem cells.


Subject(s)
Mouse Embryonic Stem Cells , Polycomb Repressive Complex 1 , Animals , Gene Expression Regulation , Mice , Mouse Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/genetics
16.
Sci Rep ; 12(1): 13908, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35974030

ABSTRACT

Early mouse development is characterized by structural and epigenetic changes while cells progress towards differentiation. At blastocyst stage, the segregation of the three primordial lineages is accompanied by establishment of differential patterns of DNA methylation and post-translational modifications of histones, such as H3K27me3. Here, we analysed the dynamics of H3K27me3 at pericentromeric heterochromatin (PCH) during early development. We also followed the localization of EZH2 and BEND3, previously shown in ESCs to drive PRC2 to hypomethylated PCH. We show that the location of H3K27me3 at PCH, in addition to H3K9me3, is a defining feature of embryonic cells in vivo. Moreover, it may play an important role in structuring PCH and preserving genomic integrity at a time of globally relaxed chromatin. At peri-implantation stages, while DNA methylation is still low, EZH2 and then H3K27me3, leave PCH in epiblast progenitors at the time of their spatial segregation from primitive endoderm cells, while BEND3 remains there up to implantation. The comparison with stem cells (ESCs and TSCs) reveals that the epigenetic marks (i.e. H3K9me3 and H3K27me3) of PCH are reset during in vitro derivation and only partially restored thereafter. This highlights possible divergences between in vitro and "in embryo" epigenetic regulation regarding constitutive heterochromatin.


Subject(s)
Heterochromatin , Histones , Animals , Blastocyst/metabolism , DNA Methylation , Epigenesis, Genetic , Heterochromatin/metabolism , Histones/metabolism , Mice
17.
Nat Cell Biol ; 24(6): 858-871, 2022 06.
Article in English | MEDLINE | ID: mdl-35697783

ABSTRACT

Human naive pluripotent stem cells have unrestricted lineage potential. Underpinning this property, naive cells are thought to lack chromatin-based lineage barriers. However, this assumption has not been tested. Here we define the chromatin-associated proteome, histone post-translational modifications and transcriptome of human naive and primed pluripotent stem cells. Our integrated analysis reveals differences in the relative abundance and activities of distinct chromatin modules. We identify a strong enrichment of polycomb repressive complex 2 (PRC2)-associated H3K27me3 in the chromatin of naive pluripotent stem cells and H3K27me3 enrichment at promoters of lineage-determining genes, including trophoblast regulators. PRC2 activity acts as a chromatin barrier restricting the differentiation of naive cells towards the trophoblast lineage, whereas inhibition of PRC2 promotes trophoblast-fate induction and cavity formation in human blastoids. Together, our results establish that human naive pluripotent stem cells are not epigenetically unrestricted, but instead possess chromatin mechanisms that oppose the induction of alternative cell fates.


Subject(s)
Pluripotent Stem Cells , Polycomb Repressive Complex 2 , Cell Differentiation/genetics , Chromatin/genetics , Histones/genetics , Humans , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Trophoblasts/metabolism
18.
Cell Stem Cell ; 29(5): 776-794.e13, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35523140

ABSTRACT

Human cholangiocyte organoids show great promise for regenerative therapies and in vitro modeling of bile duct development and diseases. However, the cystic organoids lack the branching morphology of intrahepatic bile ducts (IHBDs). Here, we report establishing human branching cholangiocyte organoid (BRCO) cultures. BRCOs self-organize into complex tubular structures resembling the IHBD architecture. Single-cell transcriptomics and functional analysis showed high similarity to primary cholangiocytes, and importantly, the branching growth mimics aspects of tubular development and is dependent on JAG1/NOTCH2 signaling. When applied to cholangiocarcinoma tumor organoids, the morphology changes to an in vitro morphology like primary tumors. Moreover, these branching cholangiocarcinoma organoids (BRCCAOs) better match the transcriptomic profile of primary tumors and showed increased chemoresistance to gemcitabine and cisplatin. In conclusion, BRCOs recapitulate a complex process of branching morphogenesis in vitro. This provides an improved model to study tubular formation, bile duct functionality, and associated biliary diseases.


Subject(s)
Cholangiocarcinoma , Organoids , Bile Ducts , Epithelial Cells , Humans , Transcriptome
19.
Clin Transl Med ; 11(12): e566, 2021 12.
Article in English | MEDLINE | ID: mdl-34954911

ABSTRACT

The well-established 3D organoid culture method enabled efficient expansion of cholangiocyte-like cells from intrahepatic (IHBD) and extrahepatic bile duct (EHBD) tissue biopsies. The extensive expansion capacity of these organoids enables various applications, from cholangiocyte disease modelling to bile duct tissue engineering. Recent research demonstrated the feasibility of culturing cholangiocyte organoids from bile, which was minimal-invasive collected via endoscopic retrograde pancreaticography (ERCP). However, a detailed analysis of these bile cholangiocyte organoids (BCOs) and the cellular region of origin was not yet demonstrated. In this study, we characterize BCOs and mirror them to the already established organoids initiated from IHBD- and EHBD-tissue. We demonstrate successful organoid-initiation from extrahepatic bile collected from gallbladder after resection and by ERCP or percutaneous transhepatic cholangiopathy from a variety of patients. BCOs initiated from these three sources of bile all show features similar to in vivo cholangiocytes. The regional-specific characteristics of the BCOs are reflected by the exclusive expression of regional common bile duct genes (HOXB2 and HOXB3) by ERCP-derived BCOs and gallbladder-derived BCOs expressing gallbladder-specific genes. Moreover, BCOs have limited hepatocyte-fate differentiation potential compared to intrahepatic cholangiocyte organoids. These results indicate that organoid-initiating cells in bile are likely of local (extrahepatic) origin and are not of intrahepatic origin. Regarding the functionality of organoid initiating cells in bile, we demonstrate that BCOs efficiently repopulate decellularized EHBD scaffolds and restore the monolayer of cholangiocyte-like cells in vitro. Bile samples obtained through minimally invasive procedures provide a safe and effective alternative source of cholangiocyte organoids. The shedding of (organoid-initiating) cholangiocytes in bile provides a convenient source of organoids for regenerative medicine.


Subject(s)
Bile Acids and Salts/genetics , Bile Ducts/chemistry , Organoids/chemistry , Phenotype , Adolescent , Adult , Aged , Bile Ducts/physiopathology , Female , Humans , Male , Middle Aged , Organoids/metabolism
20.
J Gen Virol ; 91(Pt 5): 1164-72, 2010 May.
Article in English | MEDLINE | ID: mdl-20089797

ABSTRACT

Variable genomic loci have been employed in a number of molecular epidemiology studies of white spot syndrome virus (WSSV), but it is unknown which loci are suitable molecular markers for determining WSSV spread on different spatiotemporal scales. Although previous work suggests that multiple introductions of WSSV occurred in central Vietnam, it is largely uncertain how WSSV was introduced and subsequently spread. Here, we evaluate five variable WSSV DNA loci as markers of virus spread on an intermediate (i.e. regional) scale, and develop a detailed and statistically supported model for the spread of WSSV. The genotypes of 17 WSSV isolates from along the coast of Vietnam--nine of which were newly characterized in this study--were analysed to obtain sufficient samples on an intermediate scale and to allow statistical analysis. Only the ORF23/24 variable region is an appropriate marker on this scale, as geographically proximate isolates show similar deletion sizes. The ORF14/15 variable region and variable-number tandem repeat (VNTR) loci are not useful as markers on this scale. ORF14/15 may be suitable for studying larger spatiotemporal scales, whereas VNTR loci are probably suitable for smaller scales. For ORF23/24, there is a clear pattern in the spatial distribution of WSSV: the smallest genomic deletions are found in central Vietnam, and larger deletions are found in the south and the north. WSSV genomic deletions tend to increase over time with virus spread in cultured shrimp, and our data are therefore congruent with the hypothesis that WSSV was introduced in central Vietnam and then radiated out.


Subject(s)
DNA, Viral/genetics , Molecular Epidemiology/methods , Polymorphism, Genetic , White spot syndrome virus 1/classification , White spot syndrome virus 1/genetics , Genotype , Vietnam
SELECTION OF CITATIONS
SEARCH DETAIL