Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Cell ; 83(17): 3041-3042, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37683607

RESUMO

Kavitha Sarma, corresponding author of "G-quadruplexes associated with R-loops promote CTCF binding" (in this issue of Molecular Cell), discusses her paper, scientific path, and mentorship experiences while also providing an insightful take on the struggles of being a woman in science.


Assuntos
Quadruplex G , Humanos , Feminino , Estruturas R-Loop
2.
Mol Cell ; 83(17): 3064-3079.e5, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37552993

RESUMO

CTCF is a critical regulator of genome architecture and gene expression that binds thousands of sites on chromatin. CTCF genomic localization is controlled by the recognition of a DNA sequence motif and regulated by DNA modifications. However, CTCF does not bind to all its potential sites in all cell types, raising the question of whether the underlying chromatin structure can regulate CTCF occupancy. Here, we report that R-loops facilitate CTCF binding through the formation of associated G-quadruplex (G4) structures. R-loops and G4s co-localize with CTCF at many genomic regions in mouse embryonic stem cells and promote CTCF binding to its cognate DNA motif in vitro. R-loop attenuation reduces CTCF binding in vivo. Deletion of a specific G4-forming motif in a gene reduces CTCF binding and alters gene expression. Conversely, chemical stabilization of G4s results in CTCF gains and accompanying alterations in chromatin organization, suggesting a pivotal role for G4 structures in reinforcing long-range genome interactions through CTCF.


Assuntos
Quadruplex G , Animais , Camundongos , Estruturas R-Loop , Fator de Ligação a CCCTC/metabolismo , Cromatina/genética , Genômica , Sítios de Ligação
3.
Nucleic Acids Res ; 50(16): 9162-9174, 2022 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-35998910

RESUMO

ATRX is a chromatin remodeler, which is mutated in ATRX syndrome, a neurodevelopmental disorder. ATRX mutations that alter histone binding or chromatin remodeling activities cluster in the PHD finger or the helicase domain respectively. Using engineered mouse embryonic stem cells that exclusively express ATRX protein with mutations in the PHD finger (PHDmut) or helicase domains (K1584R), we examine how specific ATRX mutations affect neurodifferentiation. ATRX PHDmut and K1584R proteins interact with the DAXX histone chaperone but show reduced localization to pericentromeres. Neurodifferentiation is both delayed and compromised in PHDmut and K1584R, and manifest differently from complete ATRX loss. We observe reduced enrichment of PHDmut protein to ATRX targets, while K1584R accumulates at these sites. Interestingly, ATRX mutations have distinct effects on the genome-wide localization of the polycomb repressive complex 2 (PRC2), with PHDmut and ATRX knockout showing reduced PRC2 binding at polycomb targets and K1584R showing loss at some sites and gains at others. Notably, each mutation associated with unique gene signatures, suggesting distinct pathways leading to impaired neurodifferentiation. Our results indicate that the histone binding and chromatin remodeling functions of ATRX play non-redundant roles in neurodevelopment, and when mutated lead to ATRX syndrome through separate regulatory pathways.


Assuntos
Histonas , Talassemia alfa , Animais , Camundongos , Histonas/genética , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Talassemia alfa/genética , Proteína Nuclear Ligada ao X/genética , Proteína Nuclear Ligada ao X/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Complexo Repressor Polycomb 2/metabolismo
4.
Methods Mol Biol ; 2528: 373-380, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35704205

RESUMO

R-loops are three-stranded, DNA:RNA hybrid-containing structures that form naturally throughout the genome as a consequence of transcription. Accurately determining the genomic locations and strand of origin of R-loops is critical to understanding their roles in gene regulation and disease. Here, we describe a nuclease-based protocol for genome-wide and strand-specific R-loop detection, which we term MapR. This method targets native R-loops for cleavage and release using a modified RNase H enzyme, followed by deep sequencing. An extension of the protocol, BisMapR, can additionally introduce strand specificity via non-denaturing bisulfite conversion of the R-loop's single-stranded DNA component. MapR and BisMapR identify R-loops with high resolution and low background, can be performed with low cell input, and require short experimental time.


Assuntos
Estruturas R-Loop , RNA , DNA/química , DNA/genética , Genômica , RNA/química , RNA/genética , Ribonuclease H/metabolismo
5.
Sci Adv ; 8(3): eabj8357, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-35061527

RESUMO

The production of noncanonical mRNA transcripts is associated with cell transformation. Driven by our previous findings on the sensitivity of T cell acute lymphoblastic leukemia (T-ALL) cells to SF3B1 inhibitors, we identified that SF3B1 inhibition blocks T-ALL growth in vivo with no notable associated toxicity. We also revealed protein stabilization of the U2 complex component SF3B1 via deubiquitination. Our studies showed that SF3B1 inhibition perturbs exon skipping, leading to nonsense-mediated decay and diminished levels of DNA damage response-related transcripts, such as the serine/threonine kinase CHEK2, and impaired DNA damage response. We also identified that SF3B1 inhibition leads to a general decrease in R-loop formation. We further demonstrate that clinically used SF3B1 inhibitors synergize with CHEK2 inhibitors and chemotherapeutic drugs to block leukemia growth. Our study provides the proof of principle for posttranslational regulation of splicing components and associated roles and therapeutic implications for the U2 complex in T cell leukemia.


Assuntos
Leucemia de Células T , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Homeostase , Humanos , Mutação , Fosfoproteínas/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Fatores de Processamento de RNA/genética , Fatores de Processamento de RNA/metabolismo
6.
Nat Commun ; 13(1): 53, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013239

RESUMO

R-loops are three-stranded nucleic acid structures that accumulate on chromatin in neurological diseases and cancers and contribute to genome instability. Using a proximity-dependent labeling system, we identified distinct classes of proteins that regulate R-loops in vivo through different mechanisms. We show that ATRX suppresses R-loops by interacting with RNAs and preventing R-loop formation. Our proteomics screen also discovered an unexpected enrichment for proteins containing zinc fingers and homeodomains. One of the most consistently enriched proteins was activity-dependent neuroprotective protein (ADNP), which is frequently mutated in ASD and causal in ADNP syndrome. We find that ADNP resolves R-loops in vitro and that it is necessary to suppress R-loops in vivo at its genomic targets. Furthermore, deletion of the ADNP homeodomain severely diminishes R-loop resolution activity in vitro, results in R-loop accumulation at ADNP targets, and compromises neuronal differentiation. Notably, patient-derived human induced pluripotent stem cells that contain an ADNP syndrome-causing mutation exhibit R-loop and CTCF accumulation at ADNP targets. Our findings point to a specific role for ADNP-mediated R-loop resolution in physiological and pathological neuronal function and, more broadly, to a role for zinc finger and homeodomain proteins in R-loop regulation, with important implications for developmental disorders and cancers.


Assuntos
Proteômica , Estruturas R-Loop/fisiologia , RNA/metabolismo , Animais , Diferenciação Celular , Cromatina , Células-Tronco Embrionárias , Instabilidade Genômica , Células HEK293 , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas , Camundongos , Mutação , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Estruturas R-Loop/genética , Dedos de Zinco
7.
Nat Immunol ; 23(1): 99-108, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34937926

RESUMO

Enzymes of the TET family are methylcytosine dioxygenases that undergo frequent mutational or functional inactivation in human cancers. Recurrent loss-of-function mutations in TET proteins are frequent in human diffuse large B cell lymphoma (DLBCL). Here, we investigate the role of TET proteins in B cell homeostasis and development of B cell lymphomas with features of DLBCL. We show that deletion of Tet2 and Tet3 genes in mature B cells in mice perturbs B cell homeostasis and results in spontaneous development of germinal center (GC)-derived B cell lymphomas with increased G-quadruplexes and R-loops. At a genome-wide level, G-quadruplexes and R-loops were associated with increased DNA double-strand breaks (DSBs) at immunoglobulin switch regions. Deletion of the DNA methyltransferase DNMT1 in TET-deficient B cells prevented expansion of GC B cells, diminished the accumulation of G-quadruplexes and R-loops and delayed B lymphoma development, consistent with the opposing functions of DNMT and TET enzymes in DNA methylation and demethylation. Clustered regularly interspaced short palindromic repeats (CRISPR)-mediated depletion of nucleases and helicases that regulate G-quadruplexes and R-loops decreased the viability of TET-deficient B cells. Our studies suggest a molecular mechanism by which TET loss of function might predispose to the development of B cell malignancies.


Assuntos
Linfócitos B/imunologia , Carcinogênese/imunologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/imunologia , Dioxigenases/imunologia , Homeostase/imunologia , Estruturas R-Loop/imunologia , Animais , Diferenciação Celular/imunologia , Metilação de DNA/imunologia , Quadruplex G , Centro Germinativo/imunologia , Camundongos , Camundongos Endogâmicos C57BL
8.
Nat Commun ; 12(1): 5321, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34493732

RESUMO

CARM1 is often overexpressed in human cancers including in ovarian cancer. However, therapeutic approaches based on CARM1 expression remain to be an unmet need. Cancer cells exploit adaptive responses such as the endoplasmic reticulum (ER) stress response for their survival through activating pathways such as the IRE1α/XBP1s pathway. Here, we report that CARM1-expressing ovarian cancer cells are selectively sensitive to inhibition of the IRE1α/XBP1s pathway. CARM1 regulates XBP1s target gene expression and directly interacts with XBP1s during ER stress response. Inhibition of the IRE1α/XBP1s pathway was effective against ovarian cancer in a CARM1-dependent manner both in vitro and in vivo in orthotopic and patient-derived xenograft models. In addition, IRE1α inhibitor B-I09 synergizes with immune checkpoint blockade anti-PD1 antibody in an immunocompetent CARM1-expressing ovarian cancer model. Our data show that pharmacological inhibition of the IRE1α/XBP1s pathway alone or in combination with immune checkpoint blockade represents a therapeutic strategy for CARM1-expressing cancers.


Assuntos
Carcinoma Epitelial do Ovário/terapia , Endorribonucleases/genética , Neoplasias Ovarianas/terapia , Receptor de Morte Celular Programada 1/genética , Proteínas Serina-Treonina Quinases/genética , Proteína-Arginina N-Metiltransferases/genética , Proteína 1 de Ligação a X-Box/genética , Animais , Anticorpos Monoclonais/farmacologia , Sequência de Bases , Benzopiranos/farmacologia , Carcinoma Epitelial do Ovário/genética , Carcinoma Epitelial do Ovário/imunologia , Carcinoma Epitelial do Ovário/patologia , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Endorribonucleases/antagonistas & inibidores , Endorribonucleases/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Himecromona/análogos & derivados , Himecromona/farmacologia , Inibidores de Checkpoint Imunológico , Camundongos , Terapia de Alvo Molecular , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Ligação Proteica , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/imunologia , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteína-Arginina N-Metiltransferases/imunologia , Transdução de Sinais , Proteína 1 de Ligação a X-Box/antagonistas & inibidores , Proteína 1 de Ligação a X-Box/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Elife ; 102021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33620319

RESUMO

R-loops are three-stranded nucleic acid structures with essential roles in many nuclear processes. However, their unchecked accumulation is associated with genome instability and is observed in neurodevelopmental diseases and cancers. Genome-wide profiling of R-loops in normal and diseased cells can help identify locations of pathogenic R-loops and advance efforts to attenuate them. We present an antibody-independent R-loop detection strategy, BisMapR, that combines nuclease-based R-loop isolation with non-denaturing bisulfite chemistry to produce genome-wide profiles that retain strand information. BisMapR achieves greater resolution and is faster than existing strand-specific R-loop profiling strategies. In mouse embryonic stem cells, we apply BisMapR to find that gene promoters form R-loops in both directions and uncover a subset of active enhancers that, despite being bidirectionally transcribed, form R-loops exclusively on one strand. BisMapR reveals a previously unnoticed feature of active enhancers and provides a tool to systematically examine their mechanisms in gene expression.


Assuntos
Endonucleases/metabolismo , Genoma , Estruturas R-Loop , Sulfitos/metabolismo , Transcrição Gênica , Animais , Linhagem Celular , Camundongos
10.
Nat Commun ; 11(1): 2219, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32376827

RESUMO

Heterochromatin in the eukaryotic genome is rigorously controlled by the concerted action of protein factors and RNAs. Here, we investigate the RNA binding function of ATRX, a chromatin remodeler with roles in silencing of repetitive regions of the genome and in recruitment of the polycomb repressive complex 2 (PRC2). We identify ATRX RNA binding regions (RBRs) and discover that the major ATRX RBR lies within the N-terminal region of the protein, distinct from its PHD and helicase domains. Deletion of this ATRX RBR (ATRXΔRBR) compromises ATRX interactions with RNAs in vitro and in vivo and alters its chromatin binding properties. Genome-wide studies reveal that loss of RNA interactions results in a redistribution of ATRX on chromatin. Finally, our studies identify a role for ATRX-RNA interactions in regulating PRC2 localization to a subset of polycomb target genes.


Assuntos
Cromatina/metabolismo , Complexo Repressor Polycomb 2/metabolismo , RNA/metabolismo , Proteína Nuclear Ligada ao X/genética , Animais , Montagem e Desmontagem da Cromatina/genética , Feminino , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Heterocromatina/metabolismo , Histonas/química , Histonas/metabolismo , Metilação , Camundongos , Ligação Proteica , Domínios Proteicos/genética , Proteína Nuclear Ligada ao X/metabolismo
11.
Curr Protoc Mol Biol ; 130(1): e113, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31943854

RESUMO

R-loops are abundant, RNA-containing chromatin structures that form in the genomes of both eukaryotes and prokaryotes. Devising methods to identify the precise genomic locations of R-loops is critical to understand how these structures regulate numerous cellular processes, including replication, termination, and chromosome segregation, and how their unscheduled formation results in disease. Here, we describe a new, highly sensitive, and antibody-independent method, MapR, to profile native R-loops genome wide. MapR takes advantage of the natural specificity of the RNase H enzyme to recognize DNA:RNA hybrids, a defining feature of R-loops, and combines it with a CUT&RUN approach to target, cleave, and release R-loops that can then be sequenced. MapR has low background, is faster than current R-loop detection technologies, and can be performed in any cell type without the need to generate stable cell lines. © 2020 by John Wiley & Sons, Inc.


Assuntos
Drosophila/genética , Escherichia coli/genética , Genoma , Genômica/métodos , Mamíferos/genética , Estruturas R-Loop/genética , Animais , Sequência de Bases , Linhagem Celular , Cromatina/metabolismo , DNA de Cadeia Simples/genética , RNA/genética , Ribonuclease H/genética , Ribonuclease H/metabolismo
12.
Cell Rep ; 29(5): 1369-1380.e5, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31665646

RESUMO

R-loops are three-stranded DNA:RNA hybrids that are implicated in many nuclear processes. While R-loops may have physiological roles, the formation of stable, aberrant R-loops has been observed in neurological disorders and cancers. Current methods to assess their genome-wide distribution rely on affinity purification, which is plagued by large input requirements, high noise, and poor sensitivity for dynamic R-loops. Here, we present MapR, a method that utilizes RNase H to guide micrococcal nuclease to R-loops, which are subsequently cleaved, released, and identified by sequencing. MapR detects R-loops formed at promoters and active enhancers that are likely to form transient R-loops due to the low transcriptional output of these regulatory elements and the short-lived nature of enhancer RNAs. MapR is as specific as existing techniques and more sensitive, allowing for genome-wide coverage with low input material in a fraction of the time.


Assuntos
Desoxirribonucleases/metabolismo , Genoma Humano , Estruturas R-Loop , Anticorpos/metabolismo , Linhagem Celular , Elementos Facilitadores Genéticos/genética , Células HEK293 , Humanos , Proteínas Recombinantes/metabolismo , Ribonuclease H/metabolismo , Sequências de Repetição em Tandem/genética
13.
Mol Cell ; 71(1): 103-116.e7, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-30008316

RESUMO

The control of cell fate is an epigenetic process initiated by transcription factors (TFs) that recognize DNA motifs and recruit activator complexes and transcriptional machineries to chromatin. Lineage specificity is thought to be provided solely by TF-motif pairing, while the recruited activators are passive. Here, we show that INTS13, a subunit of the Integrator complex, operates as monocytic/macrophagic differentiation factor. Integrator is a general activator of transcription at coding genes and is required for eRNA maturation. Here, we show that INTS13 functions as an independent sub-module and targets enhancers through Early Growth Response (EGR1/2) TFs and their co-factor NAB2. INTS13 binds poised monocytic enhancers eliciting chromatin looping and activation. Independent depletion of INTS13, EGR1, or NAB2 impairs monocytic differentiation of cell lines and primary human progenitors. Our data demonstrate that Integrator is not functionally homogeneous and has TF-specific regulatory potential, revealing a new enhancer regulatory axis that controls myeloid differentiation.


Assuntos
Diferenciação Celular , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Elementos Facilitadores Genéticos , Monócitos/metabolismo , Células Progenitoras Mieloides/metabolismo , Proteínas Repressoras/metabolismo , Linhagem Celular , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 2 de Resposta de Crescimento Precoce/genética , Humanos , Células Progenitoras Mieloides/citologia , Proteínas Repressoras/genética
14.
Cell ; 159(4): 869-83, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25417162

RESUMO

X chromosome inactivation (XCI) depends on the long noncoding RNA Xist and its recruitment of Polycomb Repressive Complex 2 (PRC2). PRC2 is also targeted to other sites throughout the genome to effect transcriptional repression. Using XCI as a model, we apply an unbiased proteomics approach to isolate Xist and PRC2 regulators and identified ATRX. ATRX unexpectedly functions as a high-affinity RNA-binding protein that directly interacts with RepA/Xist RNA to promote loading of PRC2 in vivo. Without ATRX, PRC2 cannot load onto Xist RNA nor spread in cis along the X chromosome. Moreover, epigenomic profiling reveals that genome-wide targeting of PRC2 depends on ATRX, as loss of ATRX leads to spatial redistribution of PRC2 and derepression of Polycomb responsive genes. Thus, ATRX is a required specificity determinant for PRC2 targeting and function.


Assuntos
DNA Helicases/metabolismo , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 2/metabolismo , RNA Longo não Codificante/metabolismo , Inativação do Cromossomo X , Animais , DNA Helicases/isolamento & purificação , Células-Tronco Embrionárias/metabolismo , Feminino , Masculino , Camundongos , Proteínas Nucleares/isolamento & purificação , Proteína Nuclear Ligada ao X
15.
Mol Cell ; 55(2): 171-85, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-24882207

RESUMO

Polycomb repressive complex 2 (PRC2) is a histone methyltransferase that is localized to thousands of mammalian genes. Though important to human disease and as a drug target, how PRC2 is recruited remains unclear. One model invokes cis-regulatory RNA. Herein, we biochemically and functionally probe PRC2's recognition of RNA using the X-inactivation model. We observe surprisingly high discriminatory capabilities. While SUZ12 and JARID2 subunits can bind RNA, EZH2 has highest affinity and is somewhat promiscuous. EED regulates the affinity of EZH2 for RNA, lending greater specificity to PRC2-RNA interactions. Intriguingly, while RNA is crucial for targeting, RNA inhibits EZH2's catalytic activity. JARID2 weakens PRC2's binding to RNA and relieves catalytic inhibition. We propose that RNA guides PRC2 to its target but inhibits its enzymatic activity until PRC2 associates with JARID2 on chromatin. Our study provides a molecular view of regulatory interactions between RNA and PRC2 at the chromatin interface.


Assuntos
Complexo Repressor Polycomb 2/química , RNA Longo não Codificante/química , Animais , Proteína Potenciadora do Homólogo 2 de Zeste , Metilação , Camundongos , Ligação Proteica , Processamento de Proteína Pós-Traducional , Subunidades Proteicas/química , Células Sf9 , Spodoptera
16.
Mol Cell ; 54(5): 777-90, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-24857549

RESUMO

The p53-regulated long noncoding RNA lincRNA-p21 has been proposed to act in trans via several mechanisms ranging from repressing genes in the p53 transcriptional network to regulating mRNA translation and protein stability. To further examine lincRNA-p21 function, we generated a conditional knockout mouse model. We find that lincRNA-p21 predominantly functions in cis to activate expression of its neighboring gene, p21. Mechanistically, we show that lincRNA-p21 acts in concert with hnRNP-K as a coactivator for p53-dependent p21 transcription. Additional phenotypes of lincRNA-p21 deficiency could be attributed to diminished p21 levels, including deregulated expression and altered chromatin state of some Polycomb target genes, a defective G1/S checkpoint, increased proliferation rates, and enhanced reprogramming efficiency. These findings indicate that lincRNA-p21 affects global gene expression and influences the p53 tumor suppressor pathway by acting in cis as a locus-restricted coactivator for p53-mediated p21 expression.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular , Proteínas do Grupo Polycomb/fisiologia , RNA Longo não Codificante/genética , Animais , Proliferação de Células , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Epigênese Genética , Camundongos , Camundongos Knockout , Ativação Transcricional , Transcriptoma
18.
Nature ; 504(7480): 465-469, 2013 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-24162848

RESUMO

The Xist long noncoding RNA (lncRNA) is essential for X-chromosome inactivation (XCI), the process by which mammals compensate for unequal numbers of sex chromosomes. During XCI, Xist coats the future inactive X chromosome (Xi) and recruits Polycomb repressive complex 2 (PRC2) to the X-inactivation centre (Xic). How Xist spreads silencing on a 150-megabases scale is unclear. Here we generate high-resolution maps of Xist binding on the X chromosome across a developmental time course using CHART-seq. In female cells undergoing XCI de novo, Xist follows a two-step mechanism, initially targeting gene-rich islands before spreading to intervening gene-poor domains. Xist is depleted from genes that escape XCI but may concentrate near escapee boundaries. Xist binding is linearly proportional to PRC2 density and H3 lysine 27 trimethylation (H3K27me3), indicating co-migration of Xist and PRC2. Interestingly, when Xist is acutely stripped off from the Xi in post-XCI cells, Xist recovers quickly within both gene-rich and gene-poor domains on a timescale of hours instead of days, indicating a previously primed Xi chromatin state. We conclude that Xist spreading takes distinct stage-specific forms. During initial establishment, Xist follows a two-step mechanism, but during maintenance, Xist spreads rapidly to both gene-rich and gene-poor regions.


Assuntos
RNA Longo não Codificante/metabolismo , Inativação do Cromossomo X , Cromossomo X/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Células-Tronco Embrionárias/metabolismo , Feminino , Fibroblastos/metabolismo , Inativação Gênica , Genes , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Metilação , Camundongos , Modelos Genéticos , RNA Longo não Codificante/genética , Cromossomo X/genética , Inativação do Cromossomo X/genética
19.
Curr Opin Genet Dev ; 22(2): 62-71, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22424802

RESUMO

Equalization of X linked gene expression is necessary in mammalian cells due to the presence of two X chromosomes in females and one in males. To achieve this, all female cells inactivate one of the two X chromosomes during development. This process, termed X chromosome inactivation (XCI), is a quintessential epigenetic phenomenon and involves a complex interplay between noncoding RNAs and protein factors. Progress in this area of study has consequently resulted in new approaches to study epigenetics and regulatory RNA function. Here we will discuss recent developments in the field that have advanced our understanding of XCI and its regulatory mechanisms.


Assuntos
Inativação do Cromossomo X , Animais , Dano ao DNA , Dosagem de Genes , Regulação da Expressão Gênica , Inativação Gênica , Humanos , RNA não Traduzido/genética
20.
Proc Natl Acad Sci U S A ; 107(51): 22196-201, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-21135235

RESUMO

A large fraction of the mammalian genome is transcribed into long noncoding RNAs. The RNAs remain largely uncharacterized as the field awaits new technologies to aid functional analysis. Here, we describe a unique use of locked nucleic acids (LNAs) for studying nuclear long noncoding RNA, an RNA subclass that has been less amenable to traditional knockdown techniques. We target LNAs at Xist RNA and show displacement from the X chromosome with fast kinetics. Xist transcript stability is not affected. By targeting different Xist regions, we identify a localization domain and show that polycomb repressive complex 2 (PRC2) is displaced together with Xist. Thus, PRC2 depends on RNA for both initial targeting to and stable association with chromatin. H3K27-trimethyl marks and gene silencing remain stable. Time-course analysis of RNA relocalization suggests that Xist and PRC2 bind to different regions of the X at the same time but do not reach saturating levels immediately. Thus, LNAs provide a tool for studying an emerging class of regulatory RNA and offer a window of opportunity to target epigenetic modifications with possible therapeutic applications.


Assuntos
Epigênese Genética/fisiologia , Histonas/metabolismo , Estabilidade de RNA/fisiologia , RNA não Traduzido/metabolismo , Proteínas Repressoras/metabolismo , Cromossomo X/metabolismo , Animais , Linhagem Celular Transformada , Técnicas de Silenciamento de Genes , Histonas/genética , Cinética , Camundongos , Proteínas do Grupo Polycomb , RNA Longo não Codificante , RNA não Traduzido/genética , Proteínas Repressoras/genética , Cromossomo X/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA