Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 171
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 84(4): 791-801.e6, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38262410

RESUMEN

In S phase, duplicating and assembling the whole genome into chromatin requires upregulation of replicative histone gene expression. Here, we explored how histone chaperones control histone production in human cells to ensure a proper link with chromatin assembly. Depletion of the ASF1 chaperone specifically decreases the pool of replicative histones both at the protein and RNA levels. The decrease in their overall expression, revealed by total RNA sequencing (RNA-seq), contrasted with the increase in nascent/newly synthesized RNAs observed by 4sU-labeled RNA-seq. Further inspection of replicative histone RNAs showed a 3' end processing defect with an increase of pre-mRNAs/unprocessed transcripts likely targeted to degradation. Collectively, these data argue for a production defect of replicative histone RNAs in ASF1-depleted cells. We discuss how this regulation of replicative histone RNA metabolism by ASF1 as a "chaperone checkpoint" fine-tunes the histone dosage to avoid unbalanced situations deleterious for cell survival.


Asunto(s)
Histonas , Proteínas de Saccharomyces cerevisiae , Humanos , Histonas/genética , Histonas/metabolismo , Chaperonas de Histonas/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , ARN/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
2.
Mol Cell ; 83(21): 3773-3786, 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37734377

RESUMEN

Histone variants provide versatility in the basic unit of chromatin, helping to define dynamic landscapes and cell fates. Maintaining genome integrity is paramount for the cell, and it is intimately linked with chromatin dynamics, assembly, and disassembly during DNA transactions such as replication, repair, recombination, and transcription. In this review, we focus on the family of H3 variants and their dynamics in space and time during the cell cycle. We review the distinct H3 variants' specific features along with their escort partners, the histone chaperones, compiled across different species to discuss their distinct importance considering evolution. We place H3 dynamics at different times during the cell cycle with the possible consequences for genome stability. Finally, we examine how their mutation and alteration impact disease. The emerging picture stresses key parameters in H3 dynamics to reflect on how when they are perturbed, they become a source of stress for genome integrity.


Asunto(s)
Cromatina , Histonas , Histonas/genética , Histonas/metabolismo , Cromatina/genética , Ciclo Celular/genética , ADN , Chaperonas de Histonas/genética
3.
Mol Cell ; 82(10): 1909-1923.e5, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35381196

RESUMEN

The lack of a consensus DNA sequence defining replication origins in mammals has led researchers to consider chromatin as a means to specify these regions. However, to date, there is no mechanistic understanding of how this could be achieved and maintained given that nucleosome disruption occurs with each fork passage and with transcription. Here, by genome-wide mapping of the de novo deposition of the histone variants H3.1 and H3.3 in human cells during S phase, we identified how their dual deposition mode ensures a stable marking with H3.3 flanked on both sides by H3.1. These H3.1/H3.3 boundaries correspond to the initiation zones of early origins. Loss of the H3.3 chaperone HIRA leads to the concomitant disruption of H3.1/H3.3 boundaries and initiation zones. We propose that the HIRA-dependent deposition of H3.3 preserves H3.1/H3.3 boundaries by protecting them from H3.1 invasion linked to fork progression, contributing to a chromatin-based definition of early replication zones.


Asunto(s)
Chaperonas de Histonas , Factores de Transcripción , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Histonas/genética , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Factores de Transcripción/metabolismo
4.
Annu Rev Biochem ; 83: 487-517, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24905786

RESUMEN

The functional organization of eukaryotic DNA into chromatin uses histones as components of its building block, the nucleosome. Histone chaperones, which are proteins that escort histones throughout their cellular life, are key actors in all facets of histone metabolism; they regulate the supply and dynamics of histones at chromatin for its assembly and disassembly. Histone chaperones can also participate in the distribution of histone variants, thereby defining distinct chromatin landscapes of importance for genome function, stability, and cell identity. Here, we discuss our current knowledge of the known histone chaperones and their histone partners, focusing on histone H3 and its variants. We then place them into an escort network that distributes these histones in various deposition pathways. Through their distinct interfaces, we show how they affect dynamics during DNA replication, DNA damage, and transcription, and how they maintain genome integrity. Finally, we discuss the importance of histone chaperones during development and describe how misregulation of the histone flow can link to disease.


Asunto(s)
Cromatina/química , Chaperonas de Histonas/química , Histonas/química , Nucleosomas/química , Animales , Proteínas de Ciclo Celular/metabolismo , ADN/química , Daño del ADN , Replicación del ADN , ADN Cruciforme/química , Histonas/metabolismo , Humanos , Unión Proteica
5.
Cell ; 155(1): 94-106, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-24074863

RESUMEN

Understanding how to recover fully functional and transcriptionally active chromatin when its integrity has been challenged by genotoxic stress is a critical issue. Here, by investigating how chromatin dynamics regulate transcriptional activity in response to DNA damage in human cells, we identify a pathway involving the histone chaperone histone regulator A (HIRA) to promote transcription restart after UVC damage. Our mechanistic studies reveal that HIRA accumulates at sites of UVC irradiation upon detection of DNA damage prior to repair and deposits newly synthesized H3.3 histones. This local action of HIRA depends on ubiquitylation events associated with damage recognition. Furthermore, we demonstrate that the early and transient function of HIRA in response to DNA damage primes chromatin for later reactivation of transcription. We propose that HIRA-dependent histone deposition serves as a chromatin bookmarking system to facilitate transcription recovery after genotoxic stress.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina , Daño del ADN/efectos de la radiación , Chaperonas de Histonas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Línea Celular Tumoral , Reparación del ADN , Células HeLa , Histonas/metabolismo , Humanos , Ubiquitinación , Rayos Ultravioleta
6.
Annu Rev Cell Dev Biol ; 30: 615-46, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25288118

RESUMEN

Within the nucleus, the interplay between lineage-specific transcription factors and chromatin dynamics defines cellular identity. Control of this interplay is necessary to properly balance stability and plasticity during the development and entire life span of multicellular organisms. Here, we present our current knowledge of the contribution of histone H3 variants to chromatin dynamics during development. We review the network of histone chaperones that governs their deposition timing and sites of incorporation and highlight how their distinct distribution impacts genome organization and function. We integrate the importance of H3 variants in the context of nuclear reprogramming and cell differentiation, and, using the centromere as a paradigm, we describe a case in which the identity of a given genomic locus is propagated across different cell types. Finally, we compare development to changes in stress and disease. Both physiological and pathological settings underline the importance of H3 dynamics for genome and chromatin integrity.


Asunto(s)
Epigénesis Genética/fisiología , Código de Histonas , Histonas/fisiología , Chaperonas Moleculares/fisiología , Secuencia de Aminoácidos , Animales , Blastocisto , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Secuencia Conservada , Epigénesis Genética/genética , Fertilización , Gametogénesis/genética , Histonas/química , Histonas/genética , Humanos , Chaperonas Moleculares/genética , Datos de Secuencia Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad de la Especie
7.
Genes Dev ; 33(11-12): 669-683, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30975723

RESUMEN

The transcriptional repression of alternative lineage genes is critical for cell fate commitment. Mechanisms by which locus-specific gene silencing is initiated and heritably maintained during cell division are not clearly understood. To study the maintenance of silent gene states, we investigated how the Cd4 gene is stably repressed in CD8+ T cells. Through CRISPR and shRNA screening, we identified the histone chaperone CAF-1 as a critical component for Cd4 repression. We found that the large subunit of CAF-1, Chaf1a, requires the N-terminal KER domain to associate with the histone deacetylases HDAC1/2 and the histone demethylase LSD1, enzymes that also participate in Cd4 silencing. When CAF-1 was lacking, Cd4 derepression was markedly enhanced in the absence of the de novo DNA methyltransferase Dnmt3a but not the maintenance DNA methyltransferase Dnmt1. In contrast to Dnmt1, Dnmt3a deficiency did not significantly alter levels of DNA methylation at the Cd4 locus. Instead, Dnmt3a deficiency sensitized CD8+ T cells to Cd4 derepression mediated by compromised functions of histone-modifying factors, including the enzymes associated with CAF-1. Thus, we propose that the heritable silencing of the Cd4 gene in CD8+ T cells exploits cooperative functions among the DNA methyltransferases, CAF-1, and histone-modifying enzymes.


Asunto(s)
Antígenos CD4/genética , Factor 1 de Ensamblaje de la Cromatina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteína 4 de Unión a Retinoblastoma/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Animales , Antígenos CD4/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Femenino , Regulación de la Expresión Génica , Silenciador del Gen , Chaperonas de Histonas/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Masculino , Ratones , Dominios Proteicos
8.
Nature ; 587(7834): 377-386, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32894860

RESUMEN

Here we describe the LifeTime Initiative, which aims to track, understand and target human cells during the onset and progression of complex diseases, and to analyse their response to therapy at single-cell resolution. This mission will be implemented through the development, integration and application of single-cell multi-omics and imaging, artificial intelligence and patient-derived experimental disease models during the progression from health to disease. The analysis of large molecular and clinical datasets will identify molecular mechanisms, create predictive computational models of disease progression, and reveal new drug targets and therapies. The timely detection and interception of disease embedded in an ethical and patient-centred vision will be achieved through interactions across academia, hospitals, patient associations, health data management systems and industry. The application of this strategy to key medical challenges in cancer, neurological and neuropsychiatric disorders, and infectious, chronic inflammatory and cardiovascular diseases at the single-cell level will usher in cell-based interceptive medicine in Europe over the next decade.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Atención a la Salud/métodos , Atención a la Salud/tendencias , Medicina/métodos , Medicina/tendencias , Patología , Análisis de la Célula Individual , Inteligencia Artificial , Atención a la Salud/ética , Atención a la Salud/normas , Diagnóstico Precoz , Educación Médica , Europa (Continente) , Femenino , Salud , Humanos , Legislación Médica , Masculino , Medicina/normas
9.
Mol Cell ; 72(1): 112-126.e5, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30217558

RESUMEN

Maintenance of epigenetic integrity relies on coordinated recycling and partitioning of parental histones and deposition of newly synthesized histones during DNA replication. This process depends upon a poorly characterized network of histone chaperones, remodelers, and binding proteins. Here we implicate the POLE3-POLE4 subcomplex of the leading-strand polymerase, Polε, in replication-coupled nucleosome assembly through its ability to selectively bind to histones H3-H4. Using hydrogen/deuterium exchange mass spectrometry and physical mapping, we define minimal domains necessary for interaction between POLE3-POLE4 and histones H3-H4. Biochemical analyses establish that POLE3-POLE4 is a histone chaperone that promotes tetrasome formation and DNA supercoiling in vitro. In cells, POLE3-POLE4 binds both newly synthesized and parental histones, and its depletion hinders helicase unwinding and chromatin PCNA unloading and compromises coordinated parental histone retention and new histone deposition. Collectively, our study reveals that POLE3-POLE4 possesses intrinsic H3-H4 chaperone activity, which facilitates faithful nucleosome dynamics at the replication fork.


Asunto(s)
ADN Polimerasa III/genética , Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Epigénesis Genética/genética , Histonas/biosíntesis , Nucleoproteínas/genética , Cromatina/genética , ADN Polimerasa II/química , ADN Polimerasa II/genética , ADN Polimerasa III/química , Proteínas de Unión al ADN/química , Chaperonas de Histonas/química , Chaperonas de Histonas/genética , Histonas/genética , Humanos , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Nucleoproteínas/química , Nucleosomas/química , Nucleosomas/genética , Proteínas de Unión a Poli-ADP-Ribosa/química , Proteínas de Unión a Poli-ADP-Ribosa/genética , Antígeno Nuclear de Célula en Proliferación/genética , Unión Proteica
10.
Mol Cell ; 70(3): 449-461.e5, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29727617

RESUMEN

Hard-to-replicate regions of chromosomes (e.g., pericentromeres, centromeres, and telomeres) impede replication fork progression, eventually leading, in the event of replication stress, to chromosome fragility, aging, and cancer. Our knowledge of the mechanisms controlling the stability of these regions is essentially limited to telomeres, where fragility is counteracted by the shelterin proteins. Here we show that the shelterin subunit TRF2 ensures progression of the replication fork through pericentromeric heterochromatin, but not centromeric chromatin. In a process involving its N-terminal basic domain, TRF2 binds to pericentromeric Satellite III sequences during S phase, allowing the recruitment of the G-quadruplex-resolving helicase RTEL1 to facilitate fork progression. We also show that TRF2 is required for the stability of other heterochromatic regions localized throughout the genome, paving the way for future research on heterochromatic replication and its relationship with aging and cancer.


Asunto(s)
Replicación del ADN/genética , Genoma/genética , Heterocromatina/genética , Telómero/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Línea Celular Tumoral , Centrómero/genética , Cromatina/genética , ADN Helicasas/genética , G-Cuádruplex , Células HeLa , Humanos , Fase S/genética
11.
Semin Cell Dev Biol ; 135: 13-23, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35595602

RESUMEN

Histone variant H3.3 is incorporated into chromatin throughout the cell cycle and even in non-cycling cells. This histone variant marks actively transcribed chromatin regions with high nucleosome turnover, as well as silent pericentric and telomeric repetitive regions. In the past few years, significant progress has been made in our understanding of mechanisms involved in the transcription-coupled deposition of H3.3. Here we review how, during transcription, new H3.3 deposition intermingles with the fate of the old H3.3 variant and its recycling. First, we describe pathways enabling the incorporation of newly synthesized vs old H3.3 histones in the context of transcription. We then review the current knowledge concerning differences between these two H3.3 populations, focusing on their PTMs composition. Finally, we discuss the implications of H3.3 recycling for the maintenance of the transcriptional state and underline the emerging importance of H3.3 as a potent epigenetic regulator for both maintaining and switching a transcriptional state.


Asunto(s)
Cromatina , Histonas , Histonas/genética , Histonas/metabolismo , Cromatina/genética , Nucleosomas/genética
12.
Genes Dev ; 31(5): 463-480, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28356341

RESUMEN

In mammals, centromere definition involves the histone variant CENP-A (centromere protein A), deposited by its chaperone, HJURP (Holliday junction recognition protein). Alterations in this process impair chromosome segregation and genome stability, which are also compromised by p53 inactivation in cancer. Here we found that CENP-A and HJURP are transcriptionally up-regulated in p53-null human tumors. Using an established mouse embryonic fibroblast (MEF) model combining p53 inactivation with E1A or HRas-V12 oncogene expression, we reproduced a similar up-regulation of HJURP and CENP-A. We delineate functional CDE/CHR motifs within the Hjurp and Cenpa promoters and demonstrate their roles in p53-mediated repression. To assess the importance of HJURP up-regulation in transformed murine and human cells, we used a CRISPR/Cas9 approach. Remarkably, depletion of HJURP leads to distinct outcomes depending on their p53 status. Functional p53 elicits a cell cycle arrest response, whereas, in p53-null transformed cells, the absence of arrest enables the loss of HJURP to induce severe aneuploidy and, ultimately, apoptotic cell death. We thus tested the impact of HJURP depletion in pre-established allograft tumors in mice and revealed a major block of tumor progression in vivo. We discuss a model in which an "epigenetic addiction" to the HJURP chaperone represents an Achilles' heel in p53-deficient transformed cells.


Asunto(s)
Autoantígenos/metabolismo , Transformación Celular Neoplásica/genética , Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes p53/genética , Oncogenes/genética , Secuencias de Aminoácidos/genética , Animales , Autoantígenos/genética , Línea Celular , Células Cultivadas , Proteína A Centromérica , Proteínas Cromosómicas no Histona/genética , Segregación Cromosómica/genética , Proteínas de Unión al ADN/genética , Femenino , Eliminación de Gen , Inestabilidad Genómica/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales
13.
Mol Cell ; 64(1): 65-78, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27642047

RESUMEN

Chromatin integrity is critical for cell function and identity but is challenged by DNA damage. To understand how chromatin architecture and the information that it conveys are preserved or altered following genotoxic stress, we established a system for real-time tracking of parental histones, which characterize the pre-damage chromatin state. Focusing on histone H3 dynamics after local UVC irradiation in human cells, we demonstrate that parental histones rapidly redistribute around damaged regions by a dual mechanism combining chromatin opening and histone mobilization on chromatin. Importantly, parental histones almost entirely recover and mix with new histones in repairing chromatin. Our data further define a close coordination of parental histone dynamics with DNA repair progression through the damage sensor DDB2 (DNA damage-binding protein 2). We speculate that this mechanism may contribute to maintaining a memory of the original chromatin landscape and may help preserve epigenome stability in response to DNA damage.


Asunto(s)
Cromatina/efectos de la radiación , Reparación del ADN , Técnica del Anticuerpo Fluorescente/métodos , Histonas/genética , Osteoblastos/efectos de la radiación , Línea Celular Tumoral , Cromatina/química , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Daño del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Inestabilidad Genómica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Histonas/antagonistas & inhibidores , Histonas/metabolismo , Humanos , Osteoblastos/citología , Osteoblastos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Rayos Ultravioleta
14.
Genes Dev ; 30(11): 1313-26, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27284163

RESUMEN

The CENP-T/-W histone fold complex, as an integral part of the inner kinetochore, is essential for building a proper kinetochore at the centromere in order to direct chromosome segregation during mitosis. Notably, CENP-T/-W is not inherited at centromeres, and new deposition is absolutely required at each cell cycle for kinetochore function. However, the mechanisms underlying this new deposition of CENP-T/-W at centromeres are unclear. Here, we found that CENP-T deposition at centromeres is uncoupled from DNA synthesis. We identified Spt16 and SSRP1, subunits of the H2A-H2B histone chaperone facilitates chromatin transcription (FACT), as CENP-W binding partners through a proteomic screen. We found that the C-terminal region of Spt16 binds specifically to the histone fold region of CENP-T/-W. Furthermore, depletion of Spt16 impairs CENP-T and CENP-W deposition at endogenous centromeres, and site-directed targeting of Spt16 alone is sufficient to ensure local de novo CENP-T accumulation. We propose a model in which the FACT chaperone stabilizes the soluble CENP-T/-W complex in the cell and promotes dynamics of exchange, enabling CENP-T/-W deposition at centromeres.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Chaperonas de Histonas/metabolismo , Cinetocoros/metabolismo , Factores de Elongación Transcripcional/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Centrómero/metabolismo , Proteínas de Unión al ADN/genética , Células HeLa , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Unión Proteica , Dominios Proteicos , Pliegue de Proteína , Proteómica , Proteínas Recombinantes/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Elongación Transcripcional/genética
16.
Nat Rev Genet ; 18(3): 192-208, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28138144

RESUMEN

Centromeric chromatin undergoes major changes in composition and architecture during each cell cycle. These changes in specialized chromatin facilitate kinetochore formation in mitosis to ensure proper chromosome segregation. Thus, proper orchestration of centromeric chromatin dynamics during interphase, including replication in S phase, is crucial. We provide the current view concerning the centromeric architecture associated with satellite repeat sequences in mammals and its dynamics during the cell cycle. We summarize the contributions of deposited histone variants and their chaperones, other centromeric components - including proteins and their post-translational modifications, and RNAs - and we link the expression and deposition timing of each component during the cell cycle. Because neocentromeres occur at ectopic sites, we highlight how cell cycle processes can go wrong, leading to neocentromere formation and potentially disease.


Asunto(s)
Ciclo Celular/genética , Centrómero/genética , Cromatina/genética , Segregación Cromosómica , Animales , Humanos
18.
Mol Cell ; 53(4): 631-44, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24530302

RESUMEN

Centromeres are essential for ensuring proper chromosome segregation in eukaryotes. Their definition relies on the presence of a centromere-specific H3 histone variant CenH3, known as CENP-A in mammals. Its overexpression in aggressive cancers raises questions concerning its effect on chromatin dynamics and contribution to tumorigenesis. We find that CenH3 overexpression in human cells leads to ectopic enrichment at sites of active histone turnover involving a heterotypic tetramer containing CenH3-H4 with H3.3-H4. Ectopic localization of this particle depends on the H3.3 chaperone DAXX rather than the dedicated CenH3 chaperone HJURP. This aberrant nucleosome occludes CTCF binding and has a minor effect on gene expression. Cells overexpressing CenH3 are more tolerant of DNA damage. Both the survival advantage and CTCF occlusion in these cells are dependent on DAXX. Our findings illustrate how changes in histone variant levels can disrupt chromatin dynamics and suggests a possible mechanism for cell resistance to anticancer treatments.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autoantígenos/metabolismo , Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/metabolismo , Factor de Unión a CCCTC , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Proteína A Centromérica , Cromatina/metabolismo , Mapeo Cromosómico , Proteínas Co-Represoras , Daño del ADN , Epítopos/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Espectrometría de Masas , Microscopía Fluorescente , Chaperonas Moleculares/metabolismo , Nucleosomas/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/metabolismo
19.
Nat Rev Mol Cell Biol ; 10(3): 192-206, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19234478

RESUMEN

Studies that concern the mechanism of DNA replication have provided a major framework for understanding genetic transmission through multiple cell cycles. Recent work has begun to gain insight into possible means to ensure the stable transmission of information beyond just DNA, and has led to the concept of epigenetic inheritance. Considering chromatin-based information, key candidates have arisen as epigenetic marks, including DNA and histone modifications, histone variants, non-histone chromatin proteins, nuclear RNA as well as higher-order chromatin organization. Understanding the dynamics and stability of these marks through the cell cycle is crucial in maintaining a given chromatin state.


Asunto(s)
Ciclo Celular/genética , Epigénesis Genética , Animales , Cromatina/genética , Metilación de ADN , Replicación del ADN/genética , Marcadores Genéticos/genética , Histonas/genética , Histonas/metabolismo , Humanos , Modelos Genéticos
20.
Nature ; 528(7581): 218-24, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26659182

RESUMEN

Cellular differentiation involves profound remodelling of chromatic landscapes, yet the mechanisms by which somatic cell identity is subsequently maintained remain incompletely understood. To further elucidate regulatory pathways that safeguard the somatic state, we performed two comprehensive RNA interference (RNAi) screens targeting chromatin factors during transcription-factor-mediated reprogramming of mouse fibroblasts to induced pluripotent stem cells (iPS cells). Subunits of the chromatin assembly factor-1 (CAF-1) complex, including Chaf1a and Chaf1b, emerged as the most prominent hits from both screens, followed by modulators of lysine sumoylation and heterochromatin maintenance. Optimal modulation of both CAF-1 and transcription factor levels increased reprogramming efficiency by several orders of magnitude and facilitated iPS cell formation in as little as 4 days. Mechanistically, CAF-1 suppression led to a more accessible chromatin structure at enhancer elements early during reprogramming. These changes were accompanied by a decrease in somatic heterochromatin domains, increased binding of Sox2 to pluripotency-specific targets and activation of associated genes. Notably, suppression of CAF-1 also enhanced the direct conversion of B cells into macrophages and fibroblasts into neurons. Together, our findings reveal the histone chaperone CAF-1 to be a novel regulator of somatic cell identity during transcription-factor-induced cell-fate transitions and provide a potential strategy to modulate cellular plasticity in a regenerative setting.


Asunto(s)
Reprogramación Celular/genética , Factor 1 de Ensamblaje de la Cromatina/metabolismo , Animales , Células Cultivadas , Cromatina/metabolismo , Factor 1 de Ensamblaje de la Cromatina/antagonistas & inhibidores , Factor 1 de Ensamblaje de la Cromatina/genética , Regulación de la Expresión Génica/genética , Heterocromatina/metabolismo , Ratones , Nucleosomas/metabolismo , Interferencia de ARN , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA