Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(1): 110-129.e31, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38181737

ABSTRACT

X chromosome inactivation (XCI) serves as a paradigm for RNA-mediated regulation of gene expression, wherein the long non-coding RNA XIST spreads across the X chromosome in cis to mediate gene silencing chromosome-wide. In female naive human pluripotent stem cells (hPSCs), XIST is in a dispersed configuration, and XCI does not occur, raising questions about XIST's function. We found that XIST spreads across the X chromosome and induces dampening of X-linked gene expression in naive hPSCs. Surprisingly, XIST also targets specific autosomal regions, where it induces repressive chromatin changes and gene expression dampening. Thereby, XIST equalizes X-linked gene dosage between male and female cells while inducing differences in autosomes. The dispersed Xist configuration and autosomal localization also occur transiently during XCI initiation in mouse PSCs. Together, our study identifies XIST as the regulator of X chromosome dampening, uncovers an evolutionarily conserved trans-acting role of XIST/Xist, and reveals a correlation between XIST/Xist dispersal and autosomal targeting.


Subject(s)
Genes, X-Linked , RNA, Long Noncoding , X Chromosome , Animals , Female , Humans , Male , Mice , Gene Silencing , RNA, Long Noncoding/genetics , X Chromosome/genetics , Pluripotent Stem Cells/metabolism
2.
Cell ; 184(23): 5775-5790.e30, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34739832

ABSTRACT

RNA, DNA, and protein molecules are highly organized within three-dimensional (3D) structures in the nucleus. Although RNA has been proposed to play a role in nuclear organization, exploring this has been challenging because existing methods cannot measure higher-order RNA and DNA contacts within 3D structures. To address this, we developed RNA & DNA SPRITE (RD-SPRITE) to comprehensively map the spatial organization of RNA and DNA. These maps reveal higher-order RNA-chromatin structures associated with three major classes of nuclear function: RNA processing, heterochromatin assembly, and gene regulation. These data demonstrate that hundreds of ncRNAs form high-concentration territories throughout the nucleus, that specific RNAs are required to recruit various regulators into these territories, and that these RNAs can shape long-range DNA contacts, heterochromatin assembly, and gene expression. These results demonstrate a mechanism where RNAs form high-concentration territories, bind to diffusible regulators, and guide them into compartments to regulate essential nuclear functions.


Subject(s)
Cell Nucleus/metabolism , RNA/metabolism , Animals , Cell Nucleus/drug effects , Chromobox Protein Homolog 5/metabolism , Chromosomes/metabolism , DNA/metabolism , DNA, Satellite/metabolism , DNA-Binding Proteins/metabolism , Dactinomycin/pharmacology , Female , Genome , HEK293 Cells , Heterochromatin/metabolism , Humans , Mice , Models, Biological , Multigene Family , RNA Polymerase II/metabolism , RNA Processing, Post-Transcriptional/drug effects , RNA Processing, Post-Transcriptional/genetics , RNA Splicing/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal/genetics , RNA-Binding Proteins/metabolism , Transcription, Genetic/drug effects
3.
Cell ; 184(25): 6174-6192.e32, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34813726

ABSTRACT

The lncRNA Xist forms ∼50 diffraction-limited foci to transcriptionally silence one X chromosome. How this small number of RNA foci and interacting proteins regulate a much larger number of X-linked genes is unknown. We show that Xist foci are locally confined, contain ∼2 RNA molecules, and nucleate supramolecular complexes (SMACs) that include many copies of the critical silencing protein SPEN. Aggregation and exchange of SMAC proteins generate local protein gradients that regulate broad, proximal chromatin regions. Partitioning of numerous SPEN molecules into SMACs is mediated by their intrinsically disordered regions and essential for transcriptional repression. Polycomb deposition via SMACs induces chromatin compaction and the increase in SMACs density around genes, which propagates silencing across the X chromosome. Our findings introduce a mechanism for functional nuclear compartmentalization whereby crowding of transcriptional and architectural regulators enables the silencing of many target genes by few RNA molecules.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Mitochondrial Proteins/metabolism , RNA, Long Noncoding/metabolism , X Chromosome/metabolism , Animals , Cell Line , Embryonic Stem Cells , Fibroblasts , Gene Silencing , Humans , Mice , Protein Binding , X Chromosome Inactivation
4.
Cell ; 168(3): 442-459.e20, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28111071

ABSTRACT

Oct4, Sox2, Klf4, and cMyc (OSKM) reprogram somatic cells to pluripotency. To gain a mechanistic understanding of their function, we mapped OSKM-binding, stage-specific transcription factors (TFs), and chromatin states in discrete reprogramming stages and performed loss- and gain-of-function experiments. We found that OSK predominantly bind active somatic enhancers early in reprogramming and immediately initiate their inactivation genome-wide by inducing the redistribution of somatic TFs away from somatic enhancers to sites elsewhere engaged by OSK, recruiting Hdac1, and repressing the somatic TF Fra1. Pluripotency enhancer selection is a stepwise process that also begins early in reprogramming through collaborative binding of OSK at sites with high OSK-motif density. Most pluripotency enhancers are selected later in the process and require OS and other pluripotency TFs. Somatic and pluripotency TFs modulate reprogramming efficiency when overexpressed by altering OSK targeting, somatic-enhancer inactivation, and pluripotency enhancer selection. Together, our data indicate that collaborative interactions among OSK and with stage-specific TFs direct both somatic-enhancer inactivation and pluripotency-enhancer selection to drive reprogramming.


Subject(s)
Cellular Reprogramming , Transcription Factors/metabolism , Animals , Chromatin/metabolism , Fibroblasts/metabolism , Histone Code , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Regulatory Elements, Transcriptional , SOXB1 Transcription Factors/metabolism , Silencer Elements, Transcriptional
5.
Cell ; 165(4): 777-9, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27153491

ABSTRACT

Our understanding of human pre-implantation development is limited by the availability of human embryos and cannot completely rely on mouse studies. Petropoulos et al. now provide an extensive transcriptome analysis of a large number of human pre-implantation embryos at single-cell resolution, revealing previously unrecognized features unique to early human development.


Subject(s)
Blastocyst , Transcriptome , Animals , Embryo Implantation , Embryonic Development , Gene Expression Profiling , Humans
7.
Genes Dev ; 36(19-20): 1079-1095, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36418052

ABSTRACT

Much has been learned about the mechanisms of action of pluripotency factors Oct4 and Sox2. However, as with other regulators of cell identity, little is known about the impact of disrupting their binding motifs in a native environment or the characteristics of genes they regulate. By quantitatively examining dynamic ranges of gene expression instead of focusing on conventional measures of differential expression, we found that Oct4 and Sox2 enhancer binding is strongly enriched near genes subject to large dynamic ranges of expression among cell types, with binding sites near these genes usually within superenhancers. Mutagenesis of representative Oct4:Sox2 motifs near such active, dynamically regulated genes revealed critical roles in transcriptional activation during reprogramming, with more limited roles in transcriptional maintenance in the pluripotent state. Furthermore, representative motifs near silent genes were critical for establishing but not maintaining the fully silent state, while genes whose transcript levels varied by smaller magnitudes among cell types were unaffected by nearby Oct4:Sox2 motifs. These results suggest that Oct4 and Sox2 directly establish both active and silent transcriptional states in pluripotent cells at a large number of genes subject to dynamic regulation during mammalian development, but are less important than expected for maintaining transcriptional states.


Subject(s)
Learning , Mammals , Animals , Transcriptional Activation , Binding Sites , Mutagenesis
8.
Cell ; 159(7): 1681-97, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25525883

ABSTRACT

Reprogramming to iPSCs resets the epigenome of somatic cells, including the reversal of X chromosome inactivation. We sought to gain insight into the steps underlying the reprogramming process by examining the means by which reprogramming leads to X chromosome reactivation (XCR). Analyzing single cells in situ, we found that hallmarks of the inactive X (Xi) change sequentially, providing a direct readout of reprogramming progression. Several epigenetic changes on the Xi occur in the inverse order of developmental X inactivation, whereas others are uncoupled from this sequence. Among the latter, DNA methylation has an extraordinary long persistence on the Xi during reprogramming, and, like Xist expression, is erased only after pluripotency genes are activated. Mechanistically, XCR requires both DNA demethylation and Xist silencing, ensuring that only cells undergoing faithful reprogramming initiate XCR. Our study defines the epigenetic state of multiple sequential reprogramming intermediates and establishes a paradigm for studying cell fate transitions during reprogramming.


Subject(s)
Cellular Reprogramming , Epigenesis, Genetic , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , X Chromosome/metabolism , Animals , Cdh1 Proteins/metabolism , DNA Methylation , Homeodomain Proteins/metabolism , Mice , Nanog Homeobox Protein , RNA, Long Noncoding/metabolism
9.
Cell ; 152(6): 1324-43, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23498940

ABSTRACT

Reprogramming to induced pluripotent stem cells (iPSCs) proceeds in a stepwise manner with reprogramming factor binding, transcription, and chromatin states changing during transitions. Evidence is emerging that epigenetic priming events early in the process may be critical for pluripotency induction later. Chromatin and its regulators are important controllers of reprogramming, and reprogramming factor levels, stoichiometry, and extracellular conditions influence the outcome. The rapid progress in characterizing reprogramming is benefiting applications of iPSCs and is already enabling the rational design of novel reprogramming factor cocktails. However, recent studies have also uncovered an epigenetic instability of the X chromosome in human iPSCs that warrants careful consideration.


Subject(s)
Cellular Reprogramming , Epigenesis, Genetic , Induced Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Chromatin/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism
10.
Mol Cell ; 73(2): 250-263.e5, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30527662

ABSTRACT

Metazoan chromosomes are sequentially partitioned into topologically associating domains (TADs) and then into smaller sub-domains. One class of sub-domains, insulated neighborhoods, are proposed to spatially sequester and insulate the enclosed genes through self-association and chromatin looping. However, it has not been determined functionally whether promoter-enhancer interactions and gene regulation are broadly restricted to within these loops. Here, we employed published datasets from murine embryonic stem cells (mESCs) to identify insulated neighborhoods that confine promoter-enhancer interactions and demarcate gene regulatory regions. To directly address the functionality of these regions, we depleted estrogen-related receptor ß (Esrrb), which binds the Mediator co-activator complex, to impair enhancers of genes within 222 insulated neighborhoods without causing mESC differentiation. Esrrb depletion reduces Mediator binding, promoter-enhancer looping, and expression of both nascent RNA and mRNA within the insulated neighborhoods without significantly affecting the flanking genes. Our data indicate that insulated neighborhoods represent functional regulons in mammalian genomes.


Subject(s)
Chromosomes, Mammalian , Enhancer Elements, Genetic , Insulator Elements , Mouse Embryonic Stem Cells/physiology , Promoter Regions, Genetic , Transcription, Genetic , Animals , Binding Sites , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Databases, Genetic , Down-Regulation , Mice , Protein Binding , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Cohesins
11.
Development ; 150(20)2023 10 15.
Article in English | MEDLINE | ID: mdl-37260407

ABSTRACT

Diet contributes to health at all stages of life, from embryonic development to old age. Nutrients, including vitamins, amino acids, lipids and sugars, have instructive roles in directing cell fate and function, maintaining stem cell populations, tissue homeostasis and alleviating the consequences of aging. This Review highlights recent findings that illuminate how common diets and specific nutrients impact cell fate decisions in healthy and disease contexts. We also draw attention to new models, technologies and resources that help to address outstanding questions in this emerging field and may lead to dietary approaches that promote healthy development and improve disease treatments.


Subject(s)
Diet , Nutrients , Female , Pregnancy , Humans , Vitamins , Cell Differentiation , Aging/physiology
12.
Chem Rev ; 124(8): 4734-4777, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38579177

ABSTRACT

This comprehensive Review delves into the chemical principles governing RNA-mediated crowding events, commonly referred to as granules or biological condensates. We explore the pivotal role played by RNA sequence, structure, and chemical modifications in these processes, uncovering their correlation with crowding phenomena under physiological conditions. Additionally, we investigate instances where crowding deviates from its intended function, leading to pathological consequences. By deepening our understanding of the delicate balance that governs molecular crowding driven by RNA and its implications for cellular homeostasis, we aim to shed light on this intriguing area of research. Our exploration extends to the methodologies employed to decipher the composition and structural intricacies of RNA granules, offering a comprehensive overview of the techniques used to characterize them, including relevant computational approaches. Through two detailed examples highlighting the significance of noncoding RNAs, NEAT1 and XIST, in the formation of phase-separated assemblies and their influence on the cellular landscape, we emphasize their crucial role in cellular organization and function. By elucidating the chemical underpinnings of RNA-mediated molecular crowding, investigating the role of modifications, structures, and composition of RNA granules, and exploring both physiological and aberrant phase separation phenomena, this Review provides a multifaceted understanding of the intriguing world of RNA-mediated biological condensates.


Subject(s)
RNA , RNA/chemistry , RNA/metabolism , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Animals , Nucleic Acid Conformation
13.
Nature ; 587(7832): 145-151, 2020 11.
Article in English | MEDLINE | ID: mdl-32908311

ABSTRACT

Nuclear compartments have diverse roles in regulating gene expression, yet the molecular forces and components that drive compartment formation remain largely unclear1. The long non-coding RNA Xist establishes an intra-chromosomal compartment by localizing at a high concentration in a territory spatially close to its transcription locus2 and binding diverse proteins3-5 to achieve X-chromosome inactivation (XCI)6,7. The XCI process therefore serves as a paradigm for understanding how RNA-mediated recruitment of various proteins induces a functional compartment. The properties of the inactive X (Xi)-compartment are known to change over time, because after initial Xist spreading and transcriptional shutoff a state is reached in which gene silencing remains stable even if Xist is turned off8. Here we show that the Xist RNA-binding proteins PTBP19, MATR310, TDP-4311 and CELF112 assemble on the multivalent E-repeat element of Xist7 and, via self-aggregation and heterotypic protein-protein interactions, form a condensate1 in the Xi. This condensate is required for gene silencing and for the anchoring of Xist to the Xi territory, and can be sustained in the absence of Xist. Notably, these E-repeat-binding proteins become essential coincident with transition to the Xist-independent XCI phase8, indicating that the condensate seeded by the E-repeat underlies the developmental switch from Xist-dependence to Xist-independence. Taken together, our data show that Xist forms the Xi compartment by seeding a heteromeric condensate that consists of ubiquitous RNA-binding proteins, revealing an unanticipated mechanism for heritable gene silencing.


Subject(s)
Gene Silencing , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , Animals , CELF1 Protein/metabolism , Cell Line , DNA-Binding Proteins/metabolism , Female , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Nuclear Matrix-Associated Proteins/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , X Chromosome Inactivation/genetics
15.
Genes Dev ; 31(3): 241-246, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28270516

ABSTRACT

Chromobox homolog 3 (Cbx3/heterochromatin protein 1γ [HP1γ]) stimulates cell differentiation, but its mechanism is unknown. We found that Cbx3 binds to gene promoters upon differentiation of murine embryonic stem cells (ESCs) to neural progenitor cells (NPCs) and recruits the Mediator subunit Med26. RNAi knockdown of either Cbx3 or Med26 inhibits neural differentiation while up-regulating genes involved in mesodermal lineage decisions. Thus, Cbx3 and Med26 together ensure the fidelity of lineage specification by enhancing the expression of neural genes and down-regulating genes specific to alternative fates.


Subject(s)
Cell Differentiation , Cell Lineage , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Stem Cells/cytology , Gene Expression Regulation , Mediator Complex/metabolism , Neural Stem Cells/cytology , Animals , Cells, Cultured , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , Embryonic Stem Cells/metabolism , Mediator Complex/genetics , Mesoderm/cytology , Mesoderm/metabolism , Mice , Neural Stem Cells/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/genetics
16.
Cell ; 136(2): 364-77, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19167336

ABSTRACT

Induced pluripotent stem (iPS) cells can be obtained from fibroblasts upon expression of Oct4, Sox2, Klf4, and c-Myc. To understand how these factors induce pluripotency, we carried out genome-wide analyses of their promoter binding and expression in iPS and partially reprogrammed cells. We find that target genes of the four factors strongly overlap in iPS and embryonic stem (ES) cells. In partially reprogrammed cells, many genes co-occupied by c-Myc and any of the other three factors already show an ES cell-like binding and expression pattern. In contrast, genes that are specifically co-bound by Oct4, Sox2, and Klf4 in ES cells and encode pluripotency regulators severely lack binding and transcriptional activation. Among the four factors, c-Myc promotes the most ES cell-like transcription pattern when expressed individually in fibroblasts. These data uncover temporal and separable contributions of the four factors during the reprogramming process and indicate that ectopic c-Myc predominantly acts before pluripotency regulators are activated.


Subject(s)
Cellular Reprogramming , Embryonic Stem Cells/cytology , Mice/metabolism , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Kruppel-Like Factor 4 , Nuclear Proteins/metabolism
17.
Mol Cell Proteomics ; 20: 100039, 2021.
Article in English | MEDLINE | ID: mdl-33476790

ABSTRACT

Deep proteome coverage in bottom-up proteomics requires peptide-level fractionation to simplify the complex peptide mixture before analysis by tandem mass spectrometry. By decreasing the number of coeluting precursor peptide ions, fractionation effectively reduces the complexity of the sample leading to higher sample coverage and reduced bias toward high-abundance precursors that are preferentially identified in data-dependent acquisition strategies. To achieve this goal, we report a bead-based off-line peptide fractionation method termed CIF or carboxylate-modified magnetic bead-based isopropanol gradient peptide fractionation. CIF is an extension of the SP3 (single-pot solid phase-enhanced sample preparation) strategy and provides an effective but complementary approach to other commonly used fractionation methods including strong cation exchange and reversed phase-based chromatography. We demonstrate that CIF is an effective offline separation strategy capable of increasing the depth of peptide analyte coverage both when used alone or as a second dimension of peptide fractionation in conjunction with high pH reversed phase. These features make it ideally suited for a wide range of proteomic applications including the affinity purification of low-abundance bait proteins.


Subject(s)
2-Propanol/chemistry , Carboxylic Acids/chemistry , Chemical Fractionation/methods , Peptides/chemistry , Proteomics/methods , Chromatography, Reverse-Phase , HEK293 Cells , Humans , Ion Exchange , Magnetic Phenomena , Peptides/metabolism , Proteome
18.
Anal Chem ; 94(46): 15939-15947, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36347042

ABSTRACT

High-field asymmetric waveform ion mobility spectrometry (FAIMS) enables gas-phase separations on a chromatographic time scale and has become a useful tool for proteomic applications. Despite its emerging utility, however, the molecular determinants underlying peptide separation by FAIMS have not been systematically investigated. Here, we characterize peptide transmission in a FAIMS device across a broad range of compensation voltages (CVs) and used machine learning to identify charge state and three-dimensional (3D) electrostatic peptide potential as major contributors to peptide intensity at a given CV. We also demonstrate that the machine learning model can be used to predict optimized CV values for peptides, which significantly improves parallel reaction monitoring workflows. Together, these data provide insight into peptide separation by FAIMS and highlight its utility in targeted proteomic applications.


Subject(s)
Ion Mobility Spectrometry , Proteomics , Proteomics/methods , Mass Spectrometry/methods , Peptides/chemistry
19.
Nat Methods ; 15(9): 732-740, 2018 09.
Article in English | MEDLINE | ID: mdl-30127506

ABSTRACT

Human embryonic stem cells (hESCs) can be captured in a primed state in which they resemble the postimplantation epiblast, or in a naive state where they resemble the preimplantation epiblast. Naive-cell-specific culture conditions allow the study of preimplantation development ex vivo but reportedly lead to chromosomal abnormalities, which compromises their utility in research and potential therapeutic applications. Although MEK inhibition is essential for the naive state, here we show that reduced MEK inhibition facilitated the establishment and maintenance of naive hESCs that retained naive-cell-specific features, including global DNA hypomethylation, HERVK expression, and two active X chromosomes. We further show that hESCs cultured under these modified conditions proliferated more rapidly; accrued fewer chromosomal abnormalities; and displayed changes in the phosphorylation levels of MAPK components, regulators of DNA damage/repair, and cell cycle. We thus provide a simple modification to current methods that can enable robust growth and reduced genomic instability in naive hESCs.


Subject(s)
Embryonic Stem Cells/metabolism , Genomic Instability , MAP Kinase Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , DNA Methylation , Embryonic Stem Cells/enzymology , Humans , Proteome , Transcriptome
20.
Nature ; 521(7551): 232-6, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25915022

ABSTRACT

Many long non-coding RNAs (lncRNAs) affect gene expression, but the mechanisms by which they act are still largely unknown. One of the best-studied lncRNAs is Xist, which is required for transcriptional silencing of one X chromosome during development in female mammals. Despite extensive efforts to define the mechanism of Xist-mediated transcriptional silencing, we still do not know any proteins required for this role. The main challenge is that there are currently no methods to comprehensively define the proteins that directly interact with a lncRNA in the cell. Here we develop a method to purify a lncRNA from cells and identify proteins interacting with it directly using quantitative mass spectrometry. We identify ten proteins that specifically associate with Xist, three of these proteins--SHARP, SAF-A and LBR--are required for Xist-mediated transcriptional silencing. We show that SHARP, which interacts with the SMRT co-repressor that activates HDAC3, is not only essential for silencing, but is also required for the exclusion of RNA polymerase II (Pol II) from the inactive X. Both SMRT and HDAC3 are also required for silencing and Pol II exclusion. In addition to silencing transcription, SHARP and HDAC3 are required for Xist-mediated recruitment of the polycomb repressive complex 2 (PRC2) across the X chromosome. Our results suggest that Xist silences transcription by directly interacting with SHARP, recruiting SMRT, activating HDAC3, and deacetylating histones to exclude Pol II across the X chromosome.


Subject(s)
Gene Silencing , Histone Deacetylases/metabolism , Mass Spectrometry/methods , Nuclear Proteins/metabolism , RNA, Long Noncoding/metabolism , Transcription, Genetic/genetics , X Chromosome/genetics , Acetylation , Animals , Cell Line , DNA-Binding Proteins , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/metabolism , Female , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Histones/metabolism , Male , Mice , Nuclear Receptor Co-Repressor 2/metabolism , Polycomb Repressive Complex 2/metabolism , Protein Binding , RNA Polymerase II/metabolism , RNA, Long Noncoding/genetics , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , X Chromosome/metabolism , X Chromosome Inactivation/genetics , Lamin B Receptor
SELECTION OF CITATIONS
SEARCH DETAIL