Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters

Publication year range
1.
Cell ; 187(3): 692-711.e26, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38262408

ABSTRACT

Transcription factors (TFs) can define distinct cellular identities despite nearly identical DNA-binding specificities. One mechanism for achieving regulatory specificity is DNA-guided TF cooperativity. Although in vitro studies suggest that it may be common, examples of such cooperativity remain scarce in cellular contexts. Here, we demonstrate how "Coordinator," a long DNA motif composed of common motifs bound by many basic helix-loop-helix (bHLH) and homeodomain (HD) TFs, uniquely defines the regulatory regions of embryonic face and limb mesenchyme. Coordinator guides cooperative and selective binding between the bHLH family mesenchymal regulator TWIST1 and a collective of HD factors associated with regional identities in the face and limb. TWIST1 is required for HD binding and open chromatin at Coordinator sites, whereas HD factors stabilize TWIST1 occupancy at Coordinator and titrate it away from HD-independent sites. This cooperativity results in the shared regulation of genes involved in cell-type and positional identities and ultimately shapes facial morphology and evolution.


Subject(s)
DNA-Binding Proteins , Embryonic Development , Transcription Factors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , DNA/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Mesoderm/metabolism , Transcription Factors/metabolism , Humans , Animals , Mice , Extremities/growth & development
2.
Nat Rev Mol Cell Biol ; 23(7): 481-497, 2022 07.
Article in English | MEDLINE | ID: mdl-35228718

ABSTRACT

Transposable elements (TEs) comprise about half of the mammalian genome. TEs often contain sequences capable of recruiting the host transcription machinery, which they use to express their own products and promote transposition. However, the regulatory sequences carried by TEs may affect host transcription long after the TEs have lost the ability to transpose. Recent advances in genome analysis and engineering have facilitated systematic interrogation of the regulatory activities of TEs. In this Review, we discuss diverse mechanisms by which TEs contribute to transcription regulation. Notably, TEs can donate enhancer and promoter sequences that influence the expression of host genes, modify 3D chromatin architecture and give rise to novel regulatory genes, including non-coding RNAs and transcription factors. We discuss how TEs spur regulatory evolution and facilitate the emergence of genetic novelties in mammalian physiology and development. By virtue of their repetitive and interspersed nature, TEs offer unique opportunities to dissect the effects of mutation and genomic context on the function and evolution of cis-regulatory elements. We argue that TE-centric studies hold the key to unlocking general principles of transcription regulation and evolution.


Subject(s)
DNA Transposable Elements , Regulatory Sequences, Nucleic Acid , Animals , DNA Transposable Elements/genetics , Evolution, Molecular , Gene Expression Regulation/genetics , Mammals/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/genetics
3.
Cell ; 178(6): 1421-1436.e24, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31491386

ABSTRACT

The developmental disorder Floating-Harbor syndrome (FHS) is caused by heterozygous truncating mutations in SRCAP, a gene encoding a chromatin remodeler mediating incorporation of histone variant H2A.Z. Here, we demonstrate that FHS-associated mutations result in loss of SRCAP nuclear localization, alter neural crest gene programs in human in vitro models and Xenopus embryos, and cause craniofacial defects. These defects are mediated by one of two H2A.Z subtypes, H2A.Z.2, whose knockdown mimics and whose overexpression rescues the FHS phenotype. Selective rescue by H2A.Z.2 is conferred by one of the three amino acid differences between the H2A.Z subtypes, S38/T38. We further show that H2A.Z.1 and H2A.Z.2 genomic occupancy patterns are qualitatively similar, but quantitatively distinct, and H2A.Z.2 incorporation at AT-rich enhancers and expression of their associated genes are both sensitized to SRCAP truncations. Altogether, our results illuminate the mechanism underlying a human syndrome and uncover selective functions of H2A.Z subtypes during development.


Subject(s)
Abnormalities, Multiple/genetics , Chromatin Assembly and Disassembly , Chromatin/metabolism , Craniofacial Abnormalities/genetics , Growth Disorders/genetics , Heart Septal Defects, Ventricular/genetics , Histones/genetics , Adenosine Triphosphatases/genetics , Amino Acid Substitution , Animals , Embryonic Stem Cells , HEK293 Cells , Humans , Mutation , Xenopus laevis
4.
Cell ; 167(5): 1170-1187, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863239

ABSTRACT

A class of cis-regulatory elements, called enhancers, play a central role in orchestrating spatiotemporally precise gene-expression programs during development. Consequently, divergence in enhancer sequence and activity is thought to be an important mediator of inter- and intra-species phenotypic variation. Here, we give an overview of emerging principles of enhancer function, current models of enhancer architecture, genomic substrates from which enhancers emerge during evolution, and the influence of three-dimensional genome organization on long-range gene regulation. We discuss intricate relationships between distinct elements within complex regulatory landscapes and consider their potential impact on specificity and robustness of transcriptional regulation.


Subject(s)
Enhancer Elements, Genetic , Evolution, Molecular , Genome-Wide Association Study , Transcription, Genetic , Blood/metabolism , Blood Cells/metabolism , Epigenomics , Hematopoiesis , Humans , Quantitative Trait Loci
5.
Mol Cell ; 83(3): 373-392, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36693380

ABSTRACT

Uncovering the cis-regulatory code that governs when and how much each gene is transcribed in a given genome and cellular state remains a central goal of biology. Here, we discuss major layers of regulation that influence how transcriptional outputs are encoded by DNA sequence and cellular context. We first discuss how transcription factors bind specific DNA sequences in a dosage-dependent and cooperative manner and then proceed to the cofactors that facilitate transcription factor function and mediate the activity of modular cis-regulatory elements such as enhancers, silencers, and promoters. We then consider the complex and poorly understood interplay of these diverse elements within regulatory landscapes and its relationships with chromatin states and nuclear organization. We propose that a mechanistically informed, quantitative model of transcriptional regulation that integrates these multiple regulatory layers will be the key to ultimately cracking the cis-regulatory code.


Subject(s)
Enhancer Elements, Genetic , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Promoter Regions, Genetic , Gene Expression Regulation , Base Sequence , Chromatin/genetics
6.
Mol Cell ; 83(9): 1446-1461.e6, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36996812

ABSTRACT

Enhancer clusters overlapping disease-associated mutations in Pierre Robin sequence (PRS) patients regulate SOX9 expression at genomic distances over 1.25 Mb. We applied optical reconstruction of chromatin architecture (ORCA) imaging to trace 3D locus topology during PRS-enhancer activation. We observed pronounced changes in locus topology between cell types. Subsequent analysis of single-chromatin fiber traces revealed that these ensemble-average differences arise through changes in the frequency of commonly sampled topologies. We further identified two CTCF-bound elements, internal to the SOX9 topologically associating domain, which promote stripe formation, are positioned near the domain's 3D geometric center, and bridge enhancer-promoter contacts in a series of chromatin loops. Ablation of these elements results in diminished SOX9 expression and altered domain-wide contacts. Polymer models with uniform loading across the domain and frequent cohesin collisions recapitulate this multi-loop, centrally clustered geometry. Together, we provide mechanistic insights into architectural stripe formation and gene regulation over ultra-long genomic ranges.


Subject(s)
Chromatin , Regulatory Sequences, Nucleic Acid , Humans , Chromatin/genetics , Promoter Regions, Genetic , Gene Expression Regulation , Genome , Cell Cycle Proteins/metabolism , Enhancer Elements, Genetic , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism
7.
Mol Cell ; 83(10): 1623-1639.e8, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37164018

ABSTRACT

The HUSH complex recognizes and silences foreign DNA such as viruses, transposons, and transgenes without prior exposure to its targets. Here, we show that endogenous targets of the HUSH complex fall into two distinct classes based on the presence or absence of H3K9me3. These classes are further distinguished by their transposon content and differential response to the loss of HUSH. A de novo genomic rearrangement at the Sox2 locus induces a switch from H3K9me3-independent to H3K9me3-associated HUSH targeting, resulting in silencing. We further demonstrate that HUSH interacts with the termination factor WDR82 and-via its component MPP8-with nascent RNA. HUSH accumulates at sites of high RNAPII occupancy including long exons and transcription termination sites in a manner dependent on WDR82 and CPSF. Together, our results uncover the functional diversity of HUSH targets and show that this vertebrate-specific complex exploits evolutionarily ancient transcription termination machinery for co-transcriptional chromatin targeting and genome surveillance.


Subject(s)
Gene Silencing , Transcription Factors , Transcription Factors/metabolism , Transcription, Genetic , Genome/genetics , RNA
8.
Cell ; 163(1): 68-83, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26365491

ABSTRACT

cis-regulatory changes play a central role in morphological divergence, yet the regulatory principles underlying emergence of human traits remain poorly understood. Here, we use epigenomic profiling from human and chimpanzee cranial neural crest cells to systematically and quantitatively annotate divergence of craniofacial cis-regulatory landscapes. Epigenomic divergence is often attributable to genetic variation within TF motifs at orthologous enhancers, with a novel motif being most predictive of activity biases. We explore properties of this cis-regulatory change, revealing the role of particular retroelements, uncovering broad clusters of species-biased enhancers near genes associated with human facial variation, and demonstrating that cis-regulatory divergence is linked to quantitative expression differences of crucial neural crest regulators. Our work provides a wealth of candidates for future evolutionary studies and demonstrates the value of "cellular anthropology," a strategy of using in-vitro-derived embryonic cell types to elucidate both fundamental and evolving mechanisms underlying morphological variation in higher primates.


Subject(s)
Epigenomics/methods , Evolution, Molecular , Genetic Enhancement , Neural Crest/cytology , Pan troglodytes/genetics , Animals , Embryo, Mammalian/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Transgenic , Neural Crest/metabolism , Species Specificity
9.
Mol Cell ; 81(5): 969-982.e13, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33482114

ABSTRACT

Many genes are regulated by multiple enhancers that often simultaneously activate their target gene. However, how individual enhancers collaborate to activate transcription is not well understood. Here, we dissect the functions and interdependencies of five enhancer elements that together activate Fgf5 expression during exit from naive murine pluripotency. Four intergenic elements form a super-enhancer, and most of the elements contribute to Fgf5 induction at distinct time points. A fifth, poised enhancer located in the first intron contributes to Fgf5 expression at every time point by amplifying overall Fgf5 expression levels. Despite low individual enhancer activity, together these elements strongly induce Fgf5 expression in a super-additive fashion that involves strong accumulation of RNA polymerase II at the intronic enhancer. Finally, we observe a strong anti-correlation between RNA polymerase II levels at enhancers and their distance to the closest promoter, and we identify candidate elements with properties similar to the intronic enhancer.


Subject(s)
Enhancer Elements, Genetic , Fibroblast Growth Factor 5/genetics , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/metabolism , Promoter Regions, Genetic , RNA Polymerase II/genetics , Animals , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Exons , Fibroblast Growth Factor 5/metabolism , Gene Knockout Techniques , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histones/genetics , Histones/metabolism , Introns , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , RNA Polymerase II/metabolism , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Transcription, Genetic , Red Fluorescent Protein
10.
Mol Cell ; 80(4): 699-711.e7, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33091336

ABSTRACT

CCCTC-binding factor (CTCF) and cohesin play critical roles in organizing mammalian genomes into topologically associating domains (TADs). Here, by combining genetic engineering with quantitative super-resolution stimulated emission depletion (STED) microscopy, we demonstrate that in living cells, CTCF forms clusters typically containing 2-8 molecules. A fraction of CTCF clusters, enriched for those with ≥3 molecules, are coupled with cohesin complexes with a characteristic physical distance suggestive of a defined molecular interaction. Acute degradation of the cohesin unloader WAPL or transcriptional inhibition (TI) result in increased CTCF clustering. Furthermore, the effect of TI on CTCF clusters is alleviated by the acute loss of the cohesin subunit SMC3. Our study provides quantitative characterization of CTCF clusters in living cells, uncovers the opposing effects of cohesin and transcription on CTCF clustering, and highlights the power of quantitative super-resolution microscopy as a tool to bridge the gap between biochemical and genomic methodologies in chromatin research.


Subject(s)
CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Stem Cells/cytology , Microscopy, Fluorescence/methods , Proteins/metabolism , Transcription, Genetic , Animals , CCCTC-Binding Factor/genetics , Cell Cycle Proteins/genetics , Cells, Cultured , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Mammalian , Embryonic Stem Cells/metabolism , Genetic Loci , Genome , Image Processing, Computer-Assisted , Mice , Proteins/genetics , Cohesins
11.
Cell ; 146(6): 1016-28, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925322

ABSTRACT

We report the identification of 67 previously undescribed histone modifications, increasing the current number of known histone marks by about 70%. We further investigated one of the marks, lysine crotonylation (Kcr), confirming that it represents an evolutionarily-conserved histone posttranslational modification. The unique structure and genomic localization of histone Kcr suggest that it is mechanistically and functionally different from histone lysine acetylation (Kac). Specifically, in both human somatic and mouse male germ cell genomes, histone Kcr marks either active promoters or potential enhancers. In male germinal cells immediately following meiosis, Kcr is enriched on sex chromosomes and specifically marks testis-specific genes, including a significant proportion of X-linked genes that escape sex chromosome inactivation in haploid cells. These results therefore dramatically extend the repertoire of histone PTM sites and designate Kcr as a specific mark of active sex chromosome-linked genes in postmeiotic male germ cells.


Subject(s)
Gene Expression Regulation , Histone Code , Animals , HeLa Cells , Histones/chemistry , Histones/metabolism , Humans , Lysine/metabolism , Male , Meiosis , Mice , Protein Processing, Post-Translational , Testis/cytology , Testis/metabolism
12.
Annu Rev Genomics Hum Genet ; 23: 383-412, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35483406

ABSTRACT

Variations in the form of the human face, which plays a role in our individual identities and societal interactions, have fascinated scientists and artists alike. Here, we review our current understanding of the genetics underlying variation in craniofacial morphology and disease-associated dysmorphology, synthesizing decades of progress on Mendelian syndromes in addition to more recent results from genome-wide association studies of human facial shape and disease risk. We also discuss the various approaches used to phenotype and quantify facial shape, which are of particular importance due to the complex, multipartite nature of the craniofacial form. We close by discussing how experimental studies have contributed and will further contribute to our understanding of human genetic variation and then proposing future directions and applications for the field.


Subject(s)
Genome-Wide Association Study , Humans , Phenotype
13.
Cell ; 141(7): 1108-10, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20602992

ABSTRACT

The MLL1 (mixed lineage leukemia 1) protein, which is often disrupted in leukemia, both activates and represses Hox genes during hematopoiesis. Now, Wang et al. (2010) demonstrate that the cyclophilin CyP33 underpins this regulatory switch by altering the state of MLL1 through cis-trans proline isomerization in the linker region between MLL1's third PHD finger and bromodomain.

14.
Nature ; 574(7779): 553-558, 2019 10.
Article in English | MEDLINE | ID: mdl-31645721

ABSTRACT

Age-associated chronic inflammation (inflammageing) is a central hallmark of ageing1, but its influence on specific cells remains largely unknown. Fibroblasts are present in most tissues and contribute to wound healing2,3. They are also the most widely used cell type for reprogramming to induced pluripotent stem (iPS) cells, a process that has implications for regenerative medicine and rejuvenation strategies4. Here we show that fibroblast cultures from old mice secrete inflammatory cytokines and exhibit increased variability in the efficiency of iPS cell reprogramming between mice. Variability between individuals is emerging as a feature of old age5-8, but the underlying mechanisms remain unknown. To identify drivers of this variability, we performed multi-omics profiling of fibroblast cultures from young and old mice that have different reprogramming efficiencies. This approach revealed that fibroblast cultures from old mice contain 'activated fibroblasts' that secrete inflammatory cytokines, and that the proportion of activated fibroblasts in a culture correlates with the reprogramming efficiency of that culture. Experiments in which conditioned medium was swapped between cultures showed that extrinsic factors secreted by activated fibroblasts underlie part of the variability between mice in reprogramming efficiency, and we have identified inflammatory cytokines, including TNF, as key contributors. Notably, old mice also exhibited variability in wound healing rate in vivo. Single-cell RNA-sequencing analysis identified distinct subpopulations of fibroblasts with different cytokine expression and signalling in the wounds of old mice with slow versus fast healing rates. Hence, a shift in fibroblast composition, and the ratio of inflammatory cytokines that they secrete, may drive the variability between mice in reprogramming in vitro and influence wound healing rate in vivo. This variability may reflect distinct stochastic ageing trajectories between individuals, and could help in developing personalized strategies to improve iPS cell generation and wound healing in elderly individuals.


Subject(s)
Aging/metabolism , Cellular Reprogramming , Cellular Senescence/physiology , Fibroblasts/metabolism , Wound Healing , Animals , Cell Line , Cellular Reprogramming/drug effects , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Inflammation Mediators/metabolism , Jews/genetics , Male , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA , Signal Transduction/drug effects , Single-Cell Analysis , Stochastic Processes , Time Factors , Wound Healing/drug effects
15.
Mol Cell ; 66(4): 568-576.e4, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28483418

ABSTRACT

Monomethylation of histone H3 at lysine 4 (H3K4me1) and acetylation of histone H3 at lysine 27 (H3K27ac) are correlated with transcriptionally engaged enhancer elements, but the functional impact of these modifications on enhancer activity is not well understood. Here we used CRISPR/Cas9 genome editing to separate catalytic activity-dependent and independent functions of Mll3 (Kmt2c) and Mll4 (Kmt2d, Mll2), the major enhancer H3K4 monomethyltransferases. Loss of H3K4me1 from enhancers in Mll3/4 catalytically deficient cells causes partial reduction of H3K27ac, but has surprisingly minor effects on transcription from either enhancers or promoters. In contrast, loss of Mll3/4 proteins leads to strong depletion of enhancer Pol II occupancy and eRNA synthesis, concomitant with downregulation of target genes. Interestingly, downregulated genes exhibit reduced polymerase levels in gene bodies, but not at promoters, suggestive of pause-release defects. Altogether, our results suggest that enhancer H3K4me1 provides only a minor contribution to the long-range coactivator function of Mll3/4.


Subject(s)
Embryonic Stem Cells/enzymology , Enhancer Elements, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Promoter Regions, Genetic , RNA/biosynthesis , Transcription, Genetic , Animals , CRISPR-Cas Systems , Cell Line , Gene Editing , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/genetics , Male , Methylation , Mice , Mutation , RNA/genetics , Time Factors , Transcriptional Activation , Transfection
16.
Chemphyschem ; 25(6): e202300793, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38259120

ABSTRACT

In this paper, we report a new generation of polymeric networks as potential functional material based on changes in molecular dynamics in the solid state. The material is obtained by free radical polymerization of a diacrylate derivative bearing a steroid (stator) and a 1,4-diethynyl-phenylene-d4 fragment (rotator). Polymer research using the PALS technique complements the knowledge about nanostructural changes occurring in the system in the temperature range -115 °C - +190 °C. It indicates the presence of two types of free nanovolumes in the system and the occurrence of phase transitions. The polymer is characterized using 1 H NMR, 2 H Solid Echo NMR, ATR-FTIR and Raman spectroscopies, thermal analysis, and porosimetry. It is proved that the applied procedure leads to the formation of a novel porous organic material containing multiple molecular rotors.

17.
Chemphyschem ; 25(6): e202400184, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38488206

ABSTRACT

The front cover artwork is provided by Dr habil. Izabella Jastrzebska's group from the University of Bialystok, Poland. The image shows a polymeric network with molecular rotors (MR) as crosslinks. The MR rotation is slowed or inhibited when a molecule of stored gas is placed inside the polymer material. Read the full text of the Research Article at 10.1002/cphc.202300793.

18.
J Org Chem ; 89(3): 1648-1656, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38241473

ABSTRACT

A new synthetic route to 25-hydroxy-provitamin D3 was elaborated. The synthesis consists of direct hydroxylation at C-25 of 7-dehydrocholesterol hetero Diels-Alder adducts. The adducts were prepared by [4 + 2] cycloaddition of azadienophiles to the steroidal diene. The hydroxylation reactions of adducts were carried out with different dioxiranes or with chromyl trifluoroacetate. The byproducts of these reactions were isolated and identified. The strengths and weaknesses of hydroxylation methods with different oxidizing agents were discussed.

19.
Cell ; 137(2): 203-5, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19379684

ABSTRACT

The memory of somatic cell gene expression is reset in the germline in a process that is accompanied by dramatic changes in chromatin modifications. In this issue, Katz et al. (2009) show that the histone demethylase Lsd1/Spr-5 may participate in this resetting process in the worm, thereby preventing a decline in germ cell epigenetic stability and viability over ensuing generations.


Subject(s)
Caenorhabditis elegans/embryology , Epigenesis, Genetic , Germ Cells/cytology , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Germ Cells/metabolism , Histones/metabolism , Oxidoreductases, N-Demethylating
20.
Cell ; 139(7): 1290-302, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20064375

ABSTRACT

Polycomb Repressive Complex 2 (PRC2) regulates key developmental genes in embryonic stem (ES) cells and during development. Here we show that Jarid2/Jumonji, a protein enriched in pluripotent cells and a founding member of the Jumonji C (JmjC) domain protein family, is a PRC2 subunit in ES cells. Genome-wide ChIP-seq analyses of Jarid2, Ezh2, and Suz12 binding reveal that Jarid2 and PRC2 occupy the same genomic regions. We further show that Jarid2 promotes PRC2 recruitment to the target genes while inhibiting PRC2 histone methyltransferase activity, suggesting that it acts as a "molecular rheostat" that finely calibrates PRC2 functions at developmental genes. Using Xenopus laevis as a model we demonstrate that Jarid2 knockdown impairs the induction of gastrulation genes in blastula embryos and results in failure of differentiation. Our findings illuminate a mechanism of histone methylation regulation in pluripotent cells and during early cell-fate transitions.


Subject(s)
Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Animals , Embryonic Stem Cells/metabolism , Gene Knockdown Techniques , Humans , Mice , Mitochondria/metabolism , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , RNA/metabolism , Retinoblastoma-Binding Protein 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL