Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 405
Filtrar
1.
Biol Res ; 57(1): 22, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704609

RESUMEN

BACKGROUND: Chromatin dynamics is deeply involved in processes that require access to DNA, such as transcriptional regulation. Among the factors involved in chromatin dynamics at gene regulatory regions are general regulatory factors (GRFs). These factors contribute to establishment and maintenance of nucleosome-depleted regions (NDRs). These regions are populated by nucleosomes through histone deposition and nucleosome sliding, the latter catalyzed by a number of ATP-dependent chromatin remodeling complexes, including ISW1a. It has been observed that GRFs can act as barriers against nucleosome sliding towards NDRs. However, the relative ability of the different GRFs to hinder sliding activity is currently unknown. RESULTS: Considering this, we performed a comparative analysis for the main GRFs, with focus in their ability to modulate nucleosome sliding mediated by ISW1a. Among the GRFs tested in nucleosome remodeling assays, Rap1 was the only factor displaying the ability to hinder the activity of ISW1a. This effect requires location of the Rap1 cognate sequence on linker that becomes entry DNA in the nucleosome remodeling process. In addition, Rap1 was able to hinder nucleosome assembly in octamer transfer assays. Concurrently, Rap1 displayed the highest affinity for and longest dwell time from its target sequence, compared to the other GRFs tested. Consistently, through bioinformatics analyses of publicly available genome-wide data, we found that nucleosome occupancy and histone deposition in vivo are inversely correlated with the affinity of Rap1 for its target sequences in the genome. CONCLUSIONS: Our findings point to DNA binding affinity, residence time and location at particular translational positions relative to the nucleosome core as the key features of GRFs underlying their roles played in nucleosome sliding and assembly.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN , Nucleosomas , Nucleosomas/metabolismo , Nucleosomas/genética , Ensamble y Desensamble de Cromatina/fisiología , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Histonas/metabolismo
2.
Epigenetics Chromatin ; 15(1): 41, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36544209

RESUMEN

BACKGROUND: Regulatory elements such as promoters, enhancers, and insulators interact each other to mediate molecular processes. To capture chromatin interactions of regulatory elements, 3C-derived methods such as Hi-C and Micro-C are developed. Here, we generated and analyzed Hi-C, Micro-C, and promoter capture Micro-C datasets with different sequencing depths to study chromatin interactions of regulatory elements and nucleosome positions in human prostate cancer cells. RESULTS: Compared to Hi-C, Micro-C identifies more high-resolution loops, including ones around structural variants. By evaluating the effect of sequencing depth, we revealed that more than 2 billion reads of Micro-C are needed to detect chromatin interactions at 1 kb resolution. Moreover, we found that deep-sequencing identifies additional long-range loops that are longer than 1 Mb in distance. Furthermore, we found that more than 50% of the loops are involved in insulators while less than 10% of the loops are promoter-enhancer loops. To comprehensively capture chromatin interactions that promoters are involved in, we performed promoter capture Micro-C. Promoter capture Micro-C identifies loops near promoters with a lower amount of sequencing reads. Sequencing of 160 million reads of promoter capture Micro-C resulted in reaching a plateau of identifying loops. However, there was still a subset of promoters that are not involved in loops even after deep-sequencing. By integrating Micro-C with NOMe-seq and ChIP-seq, we found that active promoters involved in loops have a more accessible region with lower levels of DNA methylation and more highly phased nucleosomes, compared to active promoters that are not involved in loops. CONCLUSION: We determined the required sequencing depth for Micro-C and promoter capture Micro-C to generate high-resolution chromatin interaction maps and loops. We also investigated the effect of sequencing coverage of Hi-C, Micro-C, and promoter capture Micro-C on detecting chromatin loops. Our analyses suggest the presence of distinct regulatory element groups, which are differently involved in nucleosome positions and chromatin interactions. This study does not only provide valuable insights on understanding chromatin interactions of regulatory elements, but also present guidelines for designing research projects on chromatin interactions among regulatory elements.


Asunto(s)
Ensamble y Desensamble de Cromatina , Nucleosomas , Secuencias Reguladoras de Ácidos Nucleicos , Humanos , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Elementos de Facilitación Genéticos , Nucleosomas/genética , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos/genética
3.
Clin Transl Med ; 12(2): e746, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35220680

RESUMEN

BACKGROUND: Aging-associated osteoporosis is frequently seen in the elderly in clinic, but efficient managements are limited because of unclear nosogenesis. The current study aims to investigate the role of melatonin on senescent bone marrow stromal cells (BMSCs) and the underlying regulating mechanism. METHODS: Melatonin levels were tested by ELISA. Gene expression profiles were performed by RNA-sequencing, enrichment of H3K36me2 on gene promoters was analyzed by Chromatin Immunoprecipitation Sequencing (ChIP-seq), and chromatin accessibility was determined by Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq). Osteogenesis of BMSCs in vitro was measured by Alizarin Red and Alkaline Phosphatase staining, and in vivo effects of melatonin was assessed by histological staining and micro computed tomography (micro-CT) scan. Correlation of NSD2 expression and severity of senile osteoporosis patients were analyzed by Pearson correlation. RESULTS: Melatonin levels were decreased during aging in human bone marrow, accompanied by downregulation of the histone methyltransferase nuclear receptor binding SET domain protein 2 (NSD2) expression in the senescent BMSCs. Melatonin stimulated the expression of NSD2 through MT1/2-mediated signaling pathways, resulting in the rebalancing of H3K36me2 and H3K27me3 modifications to increase chromatin accessibility of the osteogenic genes, runt-related transcription factor 2 (RUNX2) and bone gamma-carboxyglutamate protein (BGLAP). Melatonin promoted osteogenesis of BMSCs in vitro, and alleviates osteoporosis progression in the aging mice. In clinic, severity of senile osteoporosis (SOP) was negatively correlated with melatonin level in bone marrow, as well as NSD2 expression in BMSCs. Similarly, melatonin remarkably enhanced osteogenic differentiation of BMSCs derived from SOP patients in vitro. CONCLUSIONS: Collectively, our study dissects previously unreported mechanistic insights into the epigenetic regulating machinery of melatonin in meliorating osteogenic differentiation of senescent BMSC, and provides evidence for application of melatonin in preventing aging-associated bone loss.


Asunto(s)
Ensamble y Desensamble de Cromatina/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/farmacología , Melatonina/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Proteínas Represoras/farmacología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Diferenciación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Modelos Animales de Enfermedad , Femenino , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Masculino , Melatonina/uso terapéutico , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL/metabolismo , Persona de Mediana Edad , Osteoblastos/fisiología , Proteínas Represoras/metabolismo
4.
Nat Struct Mol Biol ; 29(2): 121-129, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35173352

RESUMEN

Chromatin remodelers are ATP-dependent enzymes that reorganize nucleosomes within all eukaryotic genomes. Here we report a complex of the Chd1 remodeler bound to a nucleosome in a nucleotide-free state, determined by cryo-EM to 2.3 Å resolution. The remodeler stimulates the nucleosome to absorb an additional nucleotide on each strand at two different locations: on the tracking strand within the ATPase binding site and on the guide strand one helical turn from the ATPase motor. Remarkably, the additional nucleotide on the tracking strand is associated with a local transformation toward an A-form geometry, explaining how sequential ratcheting of each DNA strand occurs. The structure also reveals a histone-binding motif, ChEx, which can block opposing remodelers on the nucleosome and may allow Chd1 to participate in histone reorganization during transcription.


Asunto(s)
ADN de Hongos/química , ADN de Hongos/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Nucleosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Ensamble y Desensamble de Cromatina/fisiología , Microscopía por Crioelectrón , Proteínas de Unión al ADN/genética , Modelos Biológicos , Modelos Moleculares , Proteínas Motoras Moleculares/química , Proteínas Motoras Moleculares/metabolismo , Nucleosomas/química , Nucleótidos/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
5.
Nat Rev Genet ; 23(3): 137-153, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34608297

RESUMEN

DNA methylation (DNAme) and histone post-translational modifications (PTMs) have important roles in transcriptional regulation. Although many reports have characterized the functions of such chromatin marks in isolation, recent genome-wide studies reveal surprisingly complex interactions between them. Here, we focus on the interplay between DNAme and methylation of specific lysine residues on the histone H3 tail. We describe the impact of genetic perturbation of the relevant methyltransferases in the mouse on the landscape of chromatin marks as well as the transcriptome. In addition, we discuss the specific neurodevelopmental growth syndromes and cancers resulting from pathogenic mutations in the human orthologues of these genes. Integrating these observations underscores the fundamental importance of crosstalk between DNA and histone H3 methylation in development and disease.


Asunto(s)
Cromatina/metabolismo , Metilación de ADN/genética , Enfermedad/genética , Crecimiento y Desarrollo/genética , Animales , Ensamble y Desensamble de Cromatina/fisiología , Histonas/metabolismo , Humanos , Ratones , Procesamiento Proteico-Postraduccional
6.
Mol Cell Biol ; 42(2): e0066920, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-34898280

RESUMEN

Nucleophosmin (NPM1) is a multifunctional histone chaperone that can activate acetylation-dependent transcription from chromatin templates in vitro. p300-mediated acetylation of NPM1 has been shown to further enhance its transcription activation potential. Acetylated and total NPM1 pools are increased in oral squamous cell carcinoma. However, the role of NPM1 or its acetylated form (AcNPM1) in transcriptional regulation in cells and oral tumorigenesis is not fully elucidated. Using ChIP-seq analyses, we provide the first genome-wide profile of AcNPM1 and show that AcNPM1 is enriched at transcriptional regulatory elements. AcNPM1 co-occupies marks of active transcription at promoters and DNase I hypersensitive sites at enhancers. In addition, using a high-throughput protein interaction profiling approach, we show that NPM1 interacts with RNA Pol II, general transcription factors, mediator subunits, histone acetyltransferase complexes, and chromatin remodelers. NPM1 histone chaperone activity also contributes to its transcription activation potential. Further, NPM1 depletion leads to decreased AcNPM1 occupancy and reduced expression of genes required for proliferative, migratory and invasive potential of oral cancer cells. NPM1 depletion also abrogates the growth of orthotopic tumors in mice. Collectively, these results establish that AcNPM1 functions as a coactivator during during RNA polymerase II-driven transcription and regulates the expression of genes that promote oral tumorigenesis.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Regulación de la Expresión Génica/fisiología , Chaperonas de Histonas/metabolismo , Neoplasias de la Boca/genética , Nucleofosmina/metabolismo , Animales , Carcinogénesis/metabolismo , Carcinoma de Células Escamosas/genética , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Regulación de la Expresión Génica/genética , Histonas/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética
8.
Nat Commun ; 12(1): 4057, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34210977

RESUMEN

Chromatin remodeler ALC1 (amplification in liver cancer 1) is crucial for repairing damaged DNA. It is autoinhibited and activated by nucleosomal epitopes. However, the mechanisms by which ALC1 is regulated remain unclear. Here we report the crystal structure of human ALC1 and the cryoEM structure bound to the nucleosome. The structure shows the macro domain of ALC1 binds to lobe 2 of the ATPase motor, sequestering two elements for nucleosome recognition, explaining the autoinhibition mechanism of the enzyme. The H4 tail competes with the macro domain for lobe 2-binding, explaining the requirement for this nucleosomal epitope for ALC1 activation. A dual-arginine-anchor motif of ALC1 recognizes the acidic pocket of the nucleosome, which is critical for chromatin remodeling in vitro. Together, our findings illustrate the structures of ALC1 and shed light on its regulation mechanisms, paving the way for the discovery of drugs targeting ALC1 for the treatment of cancer.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Cristalografía por Rayos X/métodos , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/química , Neoplasias Hepáticas/patología , Nucleosomas/metabolismo , Proteínas Recombinantes/química , Células Cultivadas , ADN Helicasas/química , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Dominios Proteicos , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad
9.
Nat Genet ; 53(8): 1196-1206, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34253920

RESUMEN

To systematically define molecular features in human tumor cells that determine their degree of sensitivity to human allogeneic natural killer (NK) cells, we quantified the NK cell responsiveness of hundreds of molecularly annotated 'DNA-barcoded' solid tumor cell lines in multiplexed format and applied genome-scale CRISPR-based gene-editing screens in several solid tumor cell lines, to functionally interrogate which genes in tumor cells regulate the response to NK cells. In these orthogonal studies, NK cell-sensitive tumor cells tend to exhibit 'mesenchymal-like' transcriptional programs; high transcriptional signature for chromatin remodeling complexes; high levels of B7-H6 (NCR3LG1); and low levels of HLA-E/antigen presentation genes. Importantly, transcriptional signatures of NK cell-sensitive tumor cells correlate with immune checkpoint inhibitor (ICI) resistance in clinical samples. This study provides a comprehensive map of mechanisms regulating tumor cell responses to NK cells, with implications for future biomarker-driven applications of NK cell immunotherapies.


Asunto(s)
Citotoxicidad Inmunológica/genética , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Inhibidores de Puntos de Control Inmunológico/farmacología , Células Asesinas Naturales/fisiología , Células Alogénicas/fisiología , Animales , Antígenos B7/genética , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/fisiología , Pruebas Inmunológicas de Citotoxicidad/métodos , Citotoxicidad Inmunológica/fisiología , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Genoma Humano , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Ratones Endogámicos NOD , Ensayos Antitumor por Modelo de Xenoinjerto , Antígenos HLA-E
10.
Nat Struct Mol Biol ; 28(6): 501-511, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34117481

RESUMEN

The mammalian SWI/SNF complex, or BAF complex, has a conserved and direct role in antagonizing Polycomb-mediated repression. Yet, BAF also promotes repression by Polycomb in stem cells and cancer. How BAF both antagonizes and promotes Polycomb-mediated repression remains unknown. Here, we utilize targeted protein degradation to dissect the BAF-Polycomb axis in mouse embryonic stem cells on short timescales. We report that rapid BAF depletion redistributes Polycomb repressive complexes PRC1 and PRC2 from highly occupied domains, like Hox clusters, to weakly occupied sites normally opposed by BAF. Polycomb redistribution from highly repressed domains results in their decompaction, gain of active epigenomic features and transcriptional derepression. Surprisingly, through dose-dependent degradation of PRC1 and PRC2, we identify a conventional role for BAF in Polycomb-mediated repression, in addition to global Polycomb redistribution. These findings provide new mechanistic insight into the highly dynamic state of the Polycomb-Trithorax axis.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Represión Epigenética/fisiología , Regulación de la Expresión Génica/fisiología , Complejos Multiproteicos/fisiología , Proteínas del Grupo Polycomb/fisiología , Animales , Sistemas CRISPR-Cas , Células Cultivadas , Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/fisiología , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Represión Epigenética/genética , Edición Génica , Regulación de la Expresión Génica/genética , Genes Homeobox , Genoma , Células HEK293 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Mutación con Pérdida de Función , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteolisis , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
11.
Nat Commun ; 12(1): 1865, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33767158

RESUMEN

Pluripotent cells of the mammalian embryo undergo extensive chromatin rewiring to prepare for lineage commitment after implantation. Repressive H3K27me3, deposited by Polycomb Repressive Complex 2 (PRC2), is reallocated from large blankets in pre-implantation embryos to mark promoters of developmental genes. The regulation of this global redistribution of H3K27me3 is poorly understood. Here we report a post-translational mechanism that destabilizes PRC2 to constrict H3K27me3 during lineage commitment. Using an auxin-inducible degron system, we show that the deubiquitinase Usp9x is required for mouse embryonic stem (ES) cell self-renewal. Usp9x-high ES cells have high PRC2 levels and bear a chromatin and transcriptional signature of the pre-implantation embryo, whereas Usp9x-low ES cells resemble the post-implantation, gastrulating epiblast. We show that Usp9x interacts with, deubiquitinates and stabilizes PRC2. Deletion of Usp9x in post-implantation embryos results in the derepression of genes that normally gain H3K27me3 after gastrulation, followed by the appearance of morphological abnormalities at E9.5, pointing to a recurrent link between Usp9x and PRC2 during development. Usp9x is a marker of "stemness" and is mutated in various neurological disorders and cancers. Our results unveil a Usp9x-PRC2 regulatory axis that is critical at peri-implantation and may be redeployed in other stem cell fate transitions and disease states.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Células Madre Embrionarias de Ratones/citología , Células Madre Pluripotentes/citología , Complejo Represivo Polycomb 2/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Células Cultivadas , Cromatina/metabolismo , Femenino , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interferencia de ARN , ARN Interferente Pequeño/genética , Ubiquitina Tiolesterasa/genética
12.
Nat Chem Biol ; 17(4): 403-411, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33649601

RESUMEN

Whole-genome sequencing data mining efforts have revealed numerous histone mutations in a wide range of cancer types. These occur in all four core histones in both the tail and globular domains and remain largely uncharacterized. Here we used two high-throughput approaches, a DNA-barcoded mononucleosome library and a humanized yeast library, to profile the biochemical and cellular effects of these mutations. We identified cancer-associated mutations in the histone globular domains that enhance fundamental chromatin remodeling processes, histone exchange and nucleosome sliding, and are lethal in yeast. In mammalian cells, these mutations upregulate cancer-associated gene pathways and inhibit cellular differentiation by altering expression of lineage-specific transcription factors. This work represents a comprehensive functional analysis of the histone mutational landscape in human cancers and leads to a model in which histone mutations that perturb nucleosome remodeling may contribute to disease development and/or progression.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Histonas/genética , Neoplasias/genética , Animales , Diferenciación Celular/genética , Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Biblioteca de Genes , Humanos , Mutación/genética , Nucleosomas/genética , Unión Proteica , Dominios Proteicos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
13.
Sci Rep ; 11(1): 4195, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33602956

RESUMEN

Isw1 and Chd1 are ATP-dependent nucleosome-spacing enzymes required to establish regular arrays of phased nucleosomes near transcription start sites of yeast genes. Cells lacking both Isw1 and Chd1 have extremely disrupted chromatin, with weak phasing, irregular spacing and a propensity to form close-packed dinucleosomes. The Isw1 ATPase subunit occurs in two different remodeling complexes: ISW1a (composed of Isw1 and Ioc3) and ISW1b (composed of Isw1, Ioc2 and Ioc4). The Ioc4 subunit of ISW1b binds preferentially to the H3-K36me3 mark. Here we show that ISW1b is primarily responsible for setting nucleosome spacing and resolving close-packed dinucleosomes, whereas ISW1a plays only a minor role. ISW1b and Chd1 make additive contributions to dinucleosome resolution, such that neither enzyme is capable of resolving all dinucleosomes on its own. Loss of the Set2 H3-K36 methyltransferase partly phenocopies loss of Ioc4, resulting in increased dinucleosome levels with only a weak effect on nucleosome spacing, suggesting that Set2-mediated H3-K36 trimethylation contributes to ISW1b-mediated dinucleosome separation. The H4 tail domain is required for normal nucleosome spacing but not for dinucleosome resolution. We conclude that the nucleosome spacing and dinucleosome resolving activities of ISW1b and Chd1 are critical for normal global chromatin organisation.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Nucleosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Histonas/metabolismo
14.
Nat Genet ; 53(3): 279-287, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33558757

RESUMEN

Chromatin accessibility is a hallmark of regulatory regions, entails transcription factor (TF) binding and requires nucleosomal reorganization. However, it remains unclear how dynamic this process is. In the present study, we use small-molecule inhibition of the catalytic subunit of the mouse SWI/SNF remodeler complex to show that accessibility and reduced nucleosome presence at TF-binding sites rely on persistent activity of nucleosome remodelers. Within minutes of remodeler inhibition, accessibility and TF binding decrease. Although this is irrespective of TF function, we show that the activating TF OCT4 (POU5F1) exhibits a faster response than the repressive TF REST. Accessibility, nucleosome depletion and gene expression are rapidly restored on inhibitor removal, suggesting that accessible chromatin is regenerated continuously and in a largely cell-autonomous fashion. We postulate that TF binding to chromatin and remodeler-mediated nucleosomal removal do not represent a stable situation, but instead accessible chromatin reflects an average of a dynamic process under continued renewal.


Asunto(s)
Cromatina/metabolismo , Complejos Multiproteicos/metabolismo , Factores de Transcripción/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Sitios de Unión , Línea Celular/efectos de los fármacos , Cromatina/genética , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/fisiología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/antagonistas & inhibidores , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/genética , Histonas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Complejos Multiproteicos/efectos de los fármacos , Complejos Multiproteicos/genética , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
15.
Nat Struct Mol Biol ; 28(1): 71-80, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33288924

RESUMEN

SWI/SNF chromatin remodelers modify the position and spacing of nucleosomes and, in humans, are linked to cancer. To provide insights into the assembly and regulation of this protein family, we focused on a subcomplex of the Saccharomyces cerevisiae RSC comprising its ATPase (Sth1), the essential actin-related proteins (ARPs) Arp7 and Arp9 and the ARP-binding protein Rtt102. Cryo-EM and biochemical analyses of this subcomplex shows that ARP binding induces a helical conformation in the helicase-SANT-associated (HSA) domain of Sth1. Surprisingly, the ARP module is rotated 120° relative to the full RSC about a pivot point previously identified as a regulatory hub in Sth1, suggesting that large conformational changes are part of Sth1 regulation and RSC assembly. We also show that a conserved interaction between Sth1 and the nucleosome acidic patch enhances remodeling. As some cancer-associated mutations dysregulate rather than inactivate SWI/SNF remodelers, our insights into RSC complex regulation advance a mechanistic understanding of chromatin remodeling in disease states.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Microscopía por Crioelectrón , Proteínas Nucleares/metabolismo , Nucleosomas/metabolismo , Saccharomyces cerevisiae/genética
16.
J Mol Cell Biol ; 13(5): 335-346, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-33104190

RESUMEN

Local signaling events at synapses or axon terminals are communicated to the nucleus to elicit transcriptional responses, and thereby translate information about the external environment into internal neuronal representations. This retrograde signaling is critical to dendritic growth, synapse development, and neuronal plasticity. Here, we demonstrate that neuronal activity induces retrograde translocation and nuclear accumulation of endosomal adaptor APPL1. Disrupting the interaction of APPL1 with Importin α1 abolishes nuclear accumulation of APPL1, which in turn decreases the levels of histone acetylation. We further demonstrate that retrograde translocation of APPL1 is required for the regulation of gene transcription and then maintenance of hippocampal late-phase long-term potentiation. Thus, these results illustrate an APPL1-mediated pathway that contributes to the modulation of synaptic plasticity via coupling neuronal activity with chromatin remodeling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Hipocampo/metabolismo , Neuronas/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Endosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Células PC12 , Ratas , Transducción de Señal/fisiología , Sinapsis/metabolismo
17.
Cell Rep ; 33(7): 108395, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33207205

RESUMEN

The mammalian SWitch/Sucrose Non-Fermentable (SWI/SNF) chromatin-remodeling BAF (BRG1/BRM-associated factor) complex plays an essential role in developmental and pathological processes. We show that the deletion of Baf155, which encodes a subunit of the BAF complex, in the Tie2(+) lineage (Baf155 (CKO) leads to defects in yolk sac myeloid and definitive erythroid (EryD) lineage differentiation from erythromyeloid progenitors (EMPs). The chromatin of myeloid gene loci in Baf155 CKO EMPs is mostly inaccessible and enriched mainly by the ETS binding motif. BAF155 interacts with PU.1 and is recruited to PU.1 target gene loci together with p300 and KDM6a. Treatment of Baf155 CKO embryos with GSK126, an H3K27me2/3 methyltransferase EZH2 inhibitor, rescues myeloid lineage gene expression. This study uncovers indispensable BAF-mediated chromatin remodeling of myeloid gene loci at the EMP stage. Future studies exploiting epigenetics in the generation and application of EMP derivatives for tissue repair, regeneration, and disease are warranted.


Asunto(s)
Linaje de la Célula/fisiología , Ensamble y Desensamble de Cromatina/fisiología , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/fisiología , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética/genética , Células Eritroides/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/metabolismo , Células Mieloides/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo
18.
Nat Commun ; 11(1): 4455, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32901005

RESUMEN

Dysregulated alternative splicing (AS) driving carcinogenetic mitosis remains poorly understood. Here, we demonstrate that cancer metastasis-associated antigen 1 (MTA1), a well-known oncogenic chromatin modifier, broadly interacts and co-expresses with RBPs across cancers, contributing to cancerous mitosis-related AS. Using developed fCLIP-seq technology, we show that MTA1 binds abundant transcripts, preferentially at splicing-responsible motifs, influencing the abundance and AS pattern of target transcripts. MTA1 regulates the mRNA level and guides the AS of a series of mitosis regulators. MTA1 deletion abrogated the dynamic AS switches of variants for ATRX and MYBL2 at mitotic stage, which are relevant to mitosis-related tumorigenesis. MTA1 dysfunction causes defective mitotic arrest, leads to aberrant chromosome segregation, and results in chromosomal instability (CIN), eventually contributing to tumorigenesis. Currently, little is known about the RNA splicing during mitosis; here, we uncover that MTA1 binds transcripts and orchestrates dynamic splicing of mitosis regulators in tumorigenesis.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Mitosis/fisiología , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Empalme Alternativo , Animales , Sitios de Unión/genética , Ensamble y Desensamble de Cromatina/genética , Inestabilidad Cromosómica , Femenino , Células HCT116 , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Mitosis/genética , Neoplasias/genética , Neoplasias/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transactivadores/antagonistas & inhibidores , Transactivadores/genética
19.
Biochem Pharmacol ; 180: 114200, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32805211

RESUMEN

The eukaryotic genetic material is packaged in the form of chromatin by wrapping DNA around nucleosomes. Cells maintain chromatin in a dynamic state by utilising various ATP-dependent chromatin remodelling complexes which can induce structural transformations in the chromatin. All chromatin remodelers contain an ATP hydrolysing-DNA translocase motor which facilitates nucleosomal DNA translocation. By DNA translocation ISWI and CHD subfamily remodelers slide nucleosomes and arrange them in a regularly spaced array. While SWI/SNF subfamily remodelers evict or displace nucleosomes from chromatin, which promotes recruitment of transcription machinery and DNA repair factors on the DNA. Besides DNA translocation, ISWI, CHD and INO80 subfamily remodelers escort nucleosome organisation and editing. In this review; we discuss different mechanisms by which chromatin remodelers regulate chromatin accessibility, nucleosome assembly and nucleosome editing. We attempt to elucidate how their action mediates various cellular and developmental processes, and their deregulation leads to disease pathogenesis. We emphasised on their role in cancer progression and potential therapeutic implications of these complexes. We also described the drugs and strategies which are being developed to target different subunits of remodelling complexes, histone modifying enzymes and polycomb repressive complex. This includes ATPase inhibitors, EZH2 (enhancer of zeste homolog 2) inhibitors, BET (bromodomain and extra terminal) inhibitors, PROTAC (proteolysis targeting chimaera) and inhibitors of protein-protein interaction.


Asunto(s)
Ensamble y Desensamble de Cromatina/efectos de los fármacos , Cromatina/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/metabolismo , Adenosina Trifosfatasas/antagonistas & inhibidores , Adenosina Trifosfatasas/metabolismo , Animales , Ensamble y Desensamble de Cromatina/fisiología , Reparación del ADN/efectos de los fármacos , Reparación del ADN/fisiología , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Estructura Secundaria de Proteína , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo
20.
Leukemia ; 34(12): 3348-3358, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32694618

RESUMEN

Hematopoietic stem cell (HSC) utilizes its quiescence feature to combat exhaustion for lifetime blood cell supply. To date, how certain chromatin architecture and subsequent transcription profile permit HSC quiescence remains unclear. Here, we show an essential role of chromatin remodeler zinc finger HIT-type containing 1 (Znhit1) in maintaining HSC quiescence. We find that loss of Znhit1 leads to exhaustion of stem cell pool and impairment of hematopoietic function. Mechanically, Znhit1 determines the chromatin accessibility at distal enhancers of HSC quiescence genes, including Pten, Fstl1, and Klf4, for sustained transcription and consequent PI3K-Akt signaling inhibition. Moreover, Znhit1-Pten-PI3K-Akt axis also participates in controlling myeloid expansion and B-lymphoid specification. Our findings therefore identify a dominant role of Znhit1-mediated chromatin remodeling in preserving HSC function for hematopoietic homeostasis.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Cromatina/metabolismo , Cromatina/fisiología , Ensamble y Desensamble de Cromatina/fisiología , Hematopoyesis/fisiología , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA