Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Nature ; 628(8007): 442-449, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38538798

RESUMEN

Whereas oncogenes can potentially be inhibited with small molecules, the loss of tumour suppressors is more common and is problematic because the tumour-suppressor proteins are no longer present to be targeted. Notable examples include SMARCB1-mutant cancers, which are highly lethal malignancies driven by the inactivation of a subunit of SWI/SNF (also known as BAF) chromatin-remodelling complexes. Here, to generate mechanistic insights into the consequences of SMARCB1 mutation and to identify vulnerabilities, we contributed 14 SMARCB1-mutant cell lines to a near genome-wide CRISPR screen as part of the Cancer Dependency Map Project1-3. We report that the little-studied gene DDB1-CUL4-associated factor 5 (DCAF5) is required for the survival of SMARCB1-mutant cancers. We show that DCAF5 has a quality-control function for SWI/SNF complexes and promotes the degradation of incompletely assembled SWI/SNF complexes in the absence of SMARCB1. After depletion of DCAF5, SMARCB1-deficient SWI/SNF complexes reaccumulate, bind to target loci and restore SWI/SNF-mediated gene expression to levels that are sufficient to reverse the cancer state, including in vivo. Consequently, cancer results not from the loss of SMARCB1 function per se, but rather from DCAF5-mediated degradation of SWI/SNF complexes. These data indicate that therapeutic targeting of ubiquitin-mediated quality-control factors may effectively reverse the malignant state of some cancers driven by disruption of tumour suppressor complexes.


Asunto(s)
Complejos Multiproteicos , Mutación , Neoplasias , Proteína SMARCB1 , Animales , Femenino , Humanos , Masculino , Ratones , Línea Celular Tumoral , Sistemas CRISPR-Cas , Edición Génica , Neoplasias/genética , Neoplasias/metabolismo , Proteína SMARCB1/deficiencia , Proteína SMARCB1/genética , Proteína SMARCB1/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Proteolisis , Ubiquitina/metabolismo
3.
Nature ; 618(7963): 180-187, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37225980

RESUMEN

For cells to initiate and sustain a differentiated state, it is necessary that a 'memory' of this state is transmitted through mitosis to the daughter cells1-3. Mammalian switch/sucrose non-fermentable (SWI/SNF) complexes (also known as Brg1/Brg-associated factors, or BAF) control cell identity by modulating chromatin architecture to regulate gene expression4-7, but whether they participate in cell fate memory is unclear. Here we provide evidence that subunits of SWI/SNF act as mitotic bookmarks to safeguard cell identity during cell division. The SWI/SNF core subunits SMARCE1 and SMARCB1 are displaced from enhancers but are bound to promoters during mitosis, and we show that this binding is required for appropriate reactivation of bound genes after mitotic exit. Ablation of SMARCE1 during a single mitosis in mouse embryonic stem cells is sufficient to disrupt gene expression, impair the occupancy of several established bookmarks at a subset of their targets and cause aberrant neural differentiation. Thus, SWI/SNF subunit SMARCE1 has a mitotic bookmarking role and is essential for heritable epigenetic fidelity during transcriptional reprogramming.


Asunto(s)
Diferenciación Celular , Proteínas Cromosómicas no Histona , Epigénesis Genética , Mitosis , Animales , Ratones , Diferenciación Celular/genética , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Mitosis/genética , Proteínas Cromosómicas no Histona/deficiencia , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Subunidades de Proteína/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Elementos de Facilitación Genéticos/genética , Regiones Promotoras Genéticas/genética , División Celular/genética , Epigénesis Genética/genética
4.
Mol Cell ; 82(13): 2472-2489.e8, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35537449

RESUMEN

Disruption of antagonism between SWI/SNF chromatin remodelers and polycomb repressor complexes drives the formation of numerous cancer types. Recently, an inhibitor of the polycomb protein EZH2 was approved for the treatment of a sarcoma mutant in the SWI/SNF subunit SMARCB1, but resistance occurs. Here, we performed CRISPR screens in SMARCB1-mutant rhabdoid tumor cells to identify genetic contributors to SWI/SNF-polycomb antagonism and potential resistance mechanisms. We found that loss of the H3K36 methyltransferase NSD1 caused resistance to EZH2 inhibition. We show that NSD1 antagonizes polycomb via cooperation with SWI/SNF and identify co-occurrence of NSD1 inactivation in SWI/SNF-defective cancers, indicating in vivo relevance. We demonstrate that H3K36me2 itself has an essential role in the activation of polycomb target genes as inhibition of the H3K36me2 demethylase KDM2A restores the efficacy of EZH2 inhibition in SWI/SNF-deficient cells lacking NSD1. Together our data expand the mechanistic understanding of SWI/SNF and polycomb interplay and identify NSD1 as the key for coordinating this transcriptional control.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2 , Proteínas F-Box , N-Metiltransferasa de Histona-Lisina , Histona Demetilasas con Dominio de Jumonji , Proteínas del Grupo Polycomb , Proteína SMARCB1 , Cromatina/genética , Cromatina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Tumor Rabdoide/genética , Tumor Rabdoide/metabolismo , Tumor Rabdoide/patología , Proteína SMARCB1/genética , Proteína SMARCB1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Células Tumorales Cultivadas/metabolismo
5.
Microorganisms ; 9(9)2021 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-34576871

RESUMEN

Telomeres play important roles in safeguarding the genome. The specialized repressive chromatin that assembles at telomeres and subtelomeric domains is key to this protective role. However, in many organisms, the repetitive nature of telomeric and subtelomeric sequences has hindered research efforts. The fission yeast S. pombe has provided an important model system for dissection of chromatin biology due to the relative ease of genetic manipulation and strong conservation of important regulatory proteins with higher eukaryotes. Telomeres and the telomere-binding shelterin complex are highly conserved with mammals, as is the assembly of constitutive heterochromatin at subtelomeres. In this review, we seek to summarize recent work detailing the assembly of distinct chromatin structures within subtelomeric domains in fission yeast. These include the heterochromatic SH subtelomeric domains, the telomere-associated sequences (TAS), and ST chromatin domains that assemble highly condensed chromatin clusters called knobs. Specifically, we review new insights into the sequence of subtelomeric domains, the distinct types of chromatin that assemble on these sequences and how histone H3 K36 modifications influence these chromatin structures. We address the interplay between the subdomains of chromatin structure and how subtelomeric chromatin is influenced by both the telomere-bound shelterin complexes and by euchromatic chromatin regulators internal to the subtelomeric domain. Finally, we demonstrate that telomere clustering, which is mediated via the condensed ST chromatin knob domains, does not depend on knob assembly within these domains but on Set2, which mediates H3K36 methylation.

6.
Elife ; 102021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33522486

RESUMEN

Sequencing of cancer genomes has identified recurrent somatic mutations in histones, termed oncohistones, which are frequently poorly understood. Previously we showed that fission yeast expressing only the H3.3G34R mutant identified in aggressive pediatric glioma had reduced H3K36 trimethylation and acetylation, increased genomic instability and replicative stress, and defective homology-dependent DNA damage repair. Here we show that surprisingly distinct phenotypes result from G34V (also in glioma) and G34W (giant cell tumors of bone) mutations, differentially affecting H3K36 modifications, subtelomeric silencing, genomic stability; sensitivity to irradiation, alkylating agents, and hydroxyurea; and influencing DNA repair. In cancer, only 1 of 30 alleles encoding H3 is mutated. Whilst co-expression of wild-type H3 rescues most G34 mutant phenotypes, G34R causes dominant hydroxyurea sensitivity, homologous recombination defects, and dominant subtelomeric silencing. Together, these studies demonstrate the complexity associated with different substitutions at even a single residue in H3 and highlight the utility of genetically tractable systems for their analysis.


Asunto(s)
Histonas/metabolismo , Recombinación Homóloga , Proteínas Mutantes/metabolismo , Schizosaccharomyces/metabolismo , Reparación del ADN , Replicación del ADN , Inestabilidad Genómica , Histonas/genética , Proteínas Mutantes/genética , Schizosaccharomyces/genética
7.
Cancers (Basel) ; 11(5)2019 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-31086012

RESUMEN

In this review, we describe the attributes of histone H3 mutants identified in cancer. H3 mutants were first identified in genes encoding H3.3, in pediatric high-grade glioma, and subsequently in chondrosarcomas and giant cell tumors of bone (GCTB) in adolescents. The most heavily studied are the lysine to methionine mutants K27M and K36M, which perturb the target site for specific lysine methyltransferases and dominantly perturb methylation of corresponding lysines in other histone H3 proteins. We discuss recent progress in defining the consequences of these mutations on chromatin, including a newly emerging view of the central importance of the disruption of H3K36 modification in many distinct K to M histone mutant cancers. We also review new work exploring the role of H3.3 G34 mutants identified in pediatric glioma and GCTB. G34 is not itself post-translationally modified, but G34 mutation impinges on the modification of H3K36. Here, we ask if G34R mutation generates a new site for methylation on the histone tail. Finally, we consider evidence indicating that histone mutations might be more widespread in cancer than previously thought, and if the perceived bias towards mutation of H3.3 is real or reflects the biology of tumors in which the histone mutants were first identified.

8.
Elife ; 62017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28718400

RESUMEN

Recurrent somatic mutations of H3F3A in aggressive pediatric high-grade gliomas generate K27M or G34R/V mutant histone H3.3. H3.3-G34R/V mutants are common in tumors with mutations in p53 and ATRX, an H3.3-specific chromatin remodeler. To gain insight into the role of H3-G34R, we generated fission yeast that express only the mutant histone H3. H3-G34R specifically reduces H3K36 tri-methylation and H3K36 acetylation, and mutants show partial transcriptional overlap with set2 deletions. H3-G34R mutants exhibit genomic instability and increased replication stress, including slowed replication fork restart, although DNA replication checkpoints are functional. H3-G34R mutants are defective for DNA damage repair by homologous recombination (HR), and have altered HR protein dynamics in both damaged and untreated cells. These data suggest H3-G34R slows resolution of HR-mediated repair and that unresolved replication intermediates impair chromosome segregation. This analysis of H3-G34R mutant fission yeast provides mechanistic insight into how G34R mutation may promote genomic instability in glioma.


Asunto(s)
Replicación del ADN , Inestabilidad Genómica , Histonas/metabolismo , Recombinación Homóloga , Proteínas Mutantes/metabolismo , Schizosaccharomyces/metabolismo , Reparación del ADN , Histonas/genética , Proteínas Mutantes/genética , Mutación Missense , Schizosaccharomyces/genética
9.
Cytokine ; 91: 1-5, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27940088

RESUMEN

Vitamin A is an essential nutrient for the protection of children from respiratory tract disease. Supplementation with vitamin A is frequently prescribed in the clinical setting, in part to combat deficiencies among children in developing countries, and in part to treat respiratory infections in clinical trials. This vitamin influences immune responses via multiple, and sometimes seemingly contradictory mechanisms. For example, in separate reports, vitamin A was shown to decrease Th17 T-cell activity by downregulating IL-6, and to promote B cell production of IgA by upregulating IL-6. To explain these apparent contradictions, we evaluated the effects of retinoic acid (RA), a key metabolite of vitamin A, on cell lines of respiratory tract epithelial cells (LETs) and macrophages (MACs). When triggered with LPS or Sendai virus, a mouse respiratory pathogen, these two cell lines experienced opposing influences of RA on IL-6. Both IL-6 protein production and transcript levels were downregulated by RA in LETs, but upregulated in MACs. RA also increased transcript levels of MCP-1, GMCSF, and IL-10 in MACs, but not in LETs. Conversely, when LETs, but not MACs, were exposed to RA, there was an increase in transcripts for RARß, an RA receptor with known inhibitory effects on cell metabolism. Results help explain past discrepancies in the literature by demonstrating that the effects of RA are cell target dependent, and suggest close attention be paid to cell-specific effects in clinical trials involving vitamin A supplements.


Asunto(s)
Citocinas/biosíntesis , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos/metabolismo , Mucosa Respiratoria/metabolismo , Vitamina A/farmacología , Animales , Línea Celular Transformada , Células Epiteliales/citología , Macrófagos/citología , Ratones , Mucosa Respiratoria/citología
10.
Mol Cell ; 62(2): 207-221, 2016 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-27105116

RESUMEN

Nucleosome remodeling and deacetylation (NuRD) complexes are co-transcriptional regulators implicated in differentiation, development, and diseases. Methyl-CpG binding domain (MBD) proteins play an essential role in recruitment of NuRD complexes to their target sites in chromatin. The related SHREC complex in fission yeast drives transcriptional gene silencing in heterochromatin through cooperation with HP1 proteins. How remodeler and histone deacetylase (HDAC) cooperate within NuRD complexes remains unresolved. We determined that in SHREC the two modules occupy distant sites on the scaffold protein Clr1 and that repressive activity of SHREC can be modulated by the expression level of the HDAC-associated Clr1 domain alone. Moreover, the crystal structure of Clr2 reveals an MBD-like domain mediating recruitment of the HDAC module to heterochromatin. Thus, SHREC bi-functionality is organized in two separate modules with separate recruitment mechanisms, which work together to elicit transcriptional silencing at heterochromatic loci.


Asunto(s)
Ensamble y Desensamble de Cromatina , Silenciador del Gen , Heterocromatina/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/enzimología , Acetilación , Sitios de Unión , Islas de CpG , ADN de Hongos/metabolismo , Regulación Fúngica de la Expresión Génica , Heterocromatina/química , Heterocromatina/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/química , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Modelos Moleculares , Nucleosomas/enzimología , Nucleosomas/genética , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , ARN de Hongos/metabolismo , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/genética , Relación Estructura-Actividad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
11.
Viral Immunol ; 29(2): 132-6, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26741514

RESUMEN

Vitamin A deficiencies are common throughout the world and have a significant negative influence on immune protection against viral infections. Mouse models demonstrate that the production of IgA, a first line of defense against viruses at mucosal sites, is inhibited in the context of vitamin A deficiency. In vitro, the addition of vitamin A to activated B cells can enhance IgA expression, but downregulate IgE. Previous reports have demonstrated that vitamin A modifies cytokine patterns, and in so doing may influence antibody isotype expression by an indirect mechanism. However, we have now discovered hundreds of potential response elements among Sµ, Sɛ, and Sα switch sites within immunoglobulin heavy chain loci. These hotspots appear in both mouse and human loci and include targets for vitamin receptors and related proteins (e.g., estrogen receptors) in the nuclear receptor superfamily. Full response elements with direct repeats are relatively infrequent or absent in Sγ regions although half-sites are present. Based on these results, we pose a hypothesis that nuclear receptors have a direct effect on the immunoglobulin heavy chain class switch recombination event. We propose that vitamin A may alter S site accessibility to activation-induced deaminase and nonhomologous end-joining machinery, thereby influencing the isotype switch, antibody production, and protection against viral infections at mucosal sites.


Asunto(s)
Linfocitos B/inmunología , Cadenas Pesadas de Inmunoglobulina/genética , Elementos de Respuesta , Vitamina A/metabolismo , Animales , Sitios Genéticos , Hormonas Esteroides Gonadales/metabolismo , Humanos , Cambio de Clase de Inmunoglobulina , Ratones , Hormonas Tiroideas/metabolismo
12.
EMBO Rep ; 17(1): 79-93, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26582768

RESUMEN

Maintenance of the correct level and organisation of nucleosomes is crucial for genome function. Here, we uncover a role for a conserved bromodomain AAA-ATPase, Abo1, in the maintenance of nucleosome architecture in fission yeast. Cells lacking abo1(+) experience both a reduction and mis-positioning of nucleosomes at transcribed sequences in addition to increased intragenic transcription, phenotypes that are hallmarks of defective chromatin re-establishment behind RNA polymerase II. Abo1 is recruited to gene sequences and associates with histone H3 and the histone chaperone FACT. Furthermore, the distribution of Abo1 on chromatin is disturbed by impaired FACT function. The role of Abo1 extends to some promoters and also to silent heterochromatin. Abo1 is recruited to pericentromeric heterochromatin independently of the HP1 ortholog, Swi6, where it enforces proper nucleosome occupancy. Consequently, loss of Abo1 alleviates silencing and causes elevated chromosome mis-segregation. We suggest that Abo1 provides a histone chaperone function that maintains nucleosome architecture genome-wide.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cromatina/genética , Cromatina/metabolismo , Nucleosomas/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica , ADN Intergénico , Silenciador del Gen , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Histonas/genética , Histonas/metabolismo , Nucleosomas/genética , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/genética , Factores de Transcripción/metabolismo , Transcripción Genética
13.
PLoS One ; 10(9): e0138085, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26375390

RESUMEN

Careful regulation of the cell cycle is required for proper replication, cell division, and DNA repair. DNA damage--including that induced by many anticancer drugs--results in cell cycle delay or arrest, which can allow time for repair of DNA lesions. Although its molecular mechanism of action remains a matter of debate, the anticancer ruthenium complex KP1019 has been shown to bind DNA in biophysical assays and to damage DNA of colorectal and ovarian cancer cells in vitro. KP1019 has also been shown to induce mutations and induce cell cycle arrest in Saccharomyces cerevisiae, suggesting that budding yeast can serve as an appropriate model for characterizing the cellular response to the drug. Here we use a transcriptomic approach to verify that KP1019 induces the DNA damage response (DDR) and find that KP1019 dependent expression of HUG1 requires the Dun1 checkpoint; both consistent with KP1019 DDR in budding yeast. We observe a robust KP1019 dependent delay in cell cycle progression as measured by increase in large budded cells, 2C DNA content, and accumulation of Pds1 which functions to inhibit anaphase. Importantly, we also find that deletion of RAD9, a gene required for the DDR, blocks drug-dependent changes in cell cycle progression, thereby establishing a causal link between the DDR and phenotypes induced by KP1019. Interestingly, yeast treated with KP1019 not only delay in G2/M, but also exhibit abnormal nuclear position, wherein the nucleus spans the bud neck. This morphology correlates with short, misaligned spindles and is dependent on the dynein heavy chain gene DYN1. We find that KP1019 creates an environment where cells respond to DNA damage through nuclear (transcriptional changes) and cytoplasmic (motor protein activity) events.


Asunto(s)
Anafase/efectos de los fármacos , Biomarcadores/metabolismo , Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Indazoles/farmacología , Compuestos Organometálicos/farmacología , Saccharomyces cerevisiae/efectos de los fármacos , Western Blotting , Citometría de Flujo , Perfilación de la Expresión Génica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Compuestos de Rutenio , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Huso Acromático/efectos de los fármacos
14.
J Mol Biol ; 427(9): 1779-1796, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25724843

RESUMEN

The DEAD-box RNA helicase DDX3X is frequently mutated in pediatric medulloblastoma. We dissect how these mutants affect DDX3X function with structural, biochemical, and genetic experiments. We identify an N-terminal extension ("ATP-binding loop", ABL) that is critical for the stimulation of ATP hydrolysis by RNA. We present crystal structures suggesting that the ABL interacts dynamically with ATP and confirming that the interaction occurs in solution by NMR chemical shift perturbation and isothermal titration calorimetry. DEAD-box helicases require interaction between two conserved RecA-like helicase domains, D1 and D2 for function. We use NMR chemical shift perturbation to show that DDX3X interacts specifically with double-stranded RNA through its D1 domain, with contact mediated by residues G302 and G325. Mutants of these residues, G302V and G325E, are associated with pediatric medulloblastoma. These mutants are defective in RNA-stimulated ATP hydrolysis. We show that DDX3X complements the growth defect in a ded1 temperature-sensitive strain of Schizosaccharomyces pombe, but the cancer-associated mutants G302V and G325E do not complement and exhibit protein expression defects. Taken together, our results suggest that impaired translation of important mRNA targets by mutant DDX3X represents a key step in the development of medulloblastoma.


Asunto(s)
Adenosina Trifosfato/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Meduloblastoma/genética , Mutación/genética , ARN/genética , Sitios de Unión , Neoplasias Cerebelosas/genética , Niño , Cristalización , Cristalografía por Rayos X , ARN Helicasas DEAD-box/química , Prueba de Complementación Genética , Humanos , Hidrólisis , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Biosíntesis de Proteínas , Conformación Proteica , Schizosaccharomyces/crecimiento & desarrollo , Schizosaccharomyces/metabolismo
15.
Chromosoma ; 124(2): 177-89, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25773741

RESUMEN

Brain tumors are the most common solid tumors in children. Pediatric high-grade glioma (HGG) accounts for ∼8-12 % of these brain tumors and is a devastating disease as 70-90 % of patients die within 2 years of diagnosis. The failure to advance therapy for these children over the last 30 years is largely due to limited knowledge of the molecular basis for these tumors and a lack of disease models. Recently, sequencing of tumor cells revealed that histone H3 is frequently mutated in pediatric HGG, with up to 78 % of diffuse intrinsic pontine gliomas (DIPGs) carrying K27M and 36 % of non-brainstem gliomas carrying either K27M or G34R/V mutations. Although mutations in many chromatin modifiers have been identified in cancer, this was the first demonstration that histone mutations may be drivers of disease. Subsequent studies have identified high-frequency mutation of histone H3 to K36M in chondroblastomas and to G34W/L in giant cell tumors of bone, which are diseases of adolescents and young adults. Interestingly, the G34 mutations, the K36M mutations, and the majority of K27M mutations occur in genes encoding the replacement histone H3.3. Here, we review the peculiar characteristics of histone H3.3 and use this information as a backdrop to highlight current thinking about how the identified mutations may contribute to disease development.


Asunto(s)
Neoplasias del Tronco Encefálico/genética , Carcinogénesis/genética , Glioma/genética , Histonas/genética , Mutación , Secuencia de Aminoácidos , Animales , Neoplasias del Tronco Encefálico/diagnóstico , Niño , Glioma/diagnóstico , Humanos , Datos de Secuencia Molecular , Nucleosomas/genética , Nucleosomas/metabolismo
16.
Nat Commun ; 5: 3630, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24710217

RESUMEN

Studies of paediatric cancers have shown a high frequency of mutation across epigenetic regulators. Here we sequence 633 genes, encoding the majority of known epigenetic regulatory proteins, in over 1,000 paediatric tumours to define the landscape of somatic mutations in epigenetic regulators in paediatric cancer. Our results demonstrate a marked variation in the frequency of gene mutations across 21 different paediatric cancer subtypes, with the highest frequency of mutations detected in high-grade gliomas, T-lineage acute lymphoblastic leukaemia and medulloblastoma, and a paucity of mutations in low-grade glioma and retinoblastoma. The most frequently mutated genes are H3F3A, PHF6, ATRX, KDM6A, SMARCA4, ASXL2, CREBBP, EZH2, MLL2, USP7, ASXL1, NSD2, SETD2, SMC1A and ZMYM3. We identify novel loss-of-function mutations in the ubiquitin-specific processing protease 7 (USP7) in paediatric leukaemia, which result in decreased deubiquitination activity. Collectively, our results help to define the landscape of mutations in epigenetic regulatory genes in paediatric cancer and yield a valuable new database for investigating the role of epigenetic dysregulations in cancer.


Asunto(s)
Epigénesis Genética/genética , Regulación Neoplásica de la Expresión Génica/genética , Genes Reguladores/genética , Mutación , Neoplasias/genética , Neoplasias Encefálicas/genética , Niño , Glioma/genética , Humanos , Meduloblastoma/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Neoplasias de la Retina/genética , Retinoblastoma/genética
17.
Mol Cell Biol ; 34(11): 2046-61, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24662054

RESUMEN

Mit1 is the putative chromatin remodeling subunit of the fission yeast Snf2/histone deacetylase (HDAC) repressor complex (SHREC) and is known to repress transcription at regions of heterochromatin. However, how Mit1 modifies chromatin to silence transcription is largely unknown. Here we report that Mit1 mobilizes histone octamers in vitro and requires ATP hydrolysis and conserved chromatin tethering domains, including a previously unrecognized chromodomain, to remodel nucleosomes and silence transcription. Loss of Mit1 remodeling activity results in nucleosome depletion at specific DNA sequences that display low intrinsic affinity for the histone octamer, but its contribution to antagonizing RNA polymerase II (Pol II) access and transcription is not restricted to these sites. Genetic epistasis analyses demonstrate that SHREC subunits and the transcription-coupled Set2 histone methyltransferase, which is involved in suppression of cryptic transcription at actively transcribed regions, cooperate to silence heterochromatic transcripts. In addition, we have demonstrated that Mit1's remodeling activity contributes to SHREC function independently of Clr3's histone deacetylase activity on histone H3 K14. We propose that Mit1 is a chromatin remodeling factor that cooperates with the Clr3 histone deacetylase of SHREC and other chromatin modifiers to stabilize heterochromatin structure and to prevent access to the transcriptional machinery.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Regulación Fúngica de la Expresión Génica , Heterocromatina/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Nucleosomas/genética , Proteínas Represoras/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Secuencia de Aminoácidos , Proteínas de Ciclo Celular/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Histonas/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Datos de Secuencia Molecular , Interferencia de ARN , ARN Polimerasa II/antagonistas & inhibidores , ARN Interferente Pequeño , Proteínas Represoras/genética , Proteínas de Schizosaccharomyces pombe/genética , Transcripción Genética
18.
EMBO J ; 32(17): 2321-35, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23771057

RESUMEN

Heterochromatin assembly in fission yeast depends on the Clr4 histone methyltransferase, which targets H3K9. We show that the histone deacetylase Sir2 is required for Clr4 activity at telomeres, but acts redundantly with Clr3 histone deacetylase to maintain centromeric heterochromatin. However, Sir2 is critical for Clr4 function during de novo centromeric heterochromatin assembly. We identified new targets of Sir2 and tested if their deacetylation is necessary for Clr4-mediated heterochromatin establishment. Sir2 preferentially deacetylates H4K16Ac and H3K4Ac, but mutation of these residues to mimic acetylation did not prevent Clr4-mediated heterochromatin establishment. Sir2 also deacetylates H3K9Ac and H3K14Ac. Strains bearing H3K9 or H3K14 mutations exhibit heterochromatin defects. H3K9 mutation blocks Clr4 function, but why H3K14 mutation impacts heterochromatin was not known. Here, we demonstrate that recruitment of Clr4 to centromeres is blocked by mutation of H3K14. We suggest that Sir2 deacetylates H3K14 to target Clr4 to centromeres. Further, we demonstrate that Sir2 is critical for de novo accumulation of H3K9me2 in RNAi-deficient cells. These analyses place Sir2 and H3K14 deacetylation upstream of Clr4 recruitment during heterochromatin assembly.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Heterocromatina/metabolismo , Metiltransferasas/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Proteínas de Ciclo Celular/genética , Centrómero/metabolismo , Ensamble y Desensamble de Cromatina , Heterocromatina/genética , N-Metiltransferasa de Histona-Lisina , Histonas/metabolismo , Lisina/metabolismo , Metiltransferasas/genética , Mutación , Interferencia de ARN , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Especificidad por Sustrato , Telómero/genética , Telómero/metabolismo
19.
Mol Cell ; 47(2): 153-5, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22840999

RESUMEN

In this issue of Molecular Cell, Ishida et al. (2012) and Keller et al. (2012) show distinct outcomes for heterochromatic RNAs that bind different chromodomain proteins; Chp1 tethers transcripts to centromeres, whereas Swi6(HP1)-bound transcripts are evicted from chromatin and destroyed.

20.
Chromosome Res ; 20(5): 521-34, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22733402

RESUMEN

Distinct regions of the eukaryotic genome are packaged into different types of chromatin, with euchromatin representing gene rich, transcriptionally active regions and heterochromatin more condensed and gene poor. The assembly and maintenance of heterochromatin is important for many aspects of genome control, including silencing of gene transcription, suppression of recombination, and to ensure proper chromosome segregation. The precise mechanisms underlying heterochromatin establishment and maintenance are still unclear, but much progress has been made towards understanding this process during the last few years, particularly from studies performed in fission yeast. In this review, we hope to provide a conceptual model of centromeric heterochromatin in fission yeast that integrates our current understanding of the competing forces of transcription, replication, and RNA decay that influence its assembly and propagation.


Asunto(s)
Centrómero/metabolismo , Ensamble y Desensamble de Cromatina , Heterocromatina/metabolismo , Interferencia de ARN , Schizosaccharomyces/metabolismo , Transcripción Genética , Animales , Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Epigénesis Genética , Histonas/metabolismo , Mamíferos , Metilación , Complejos Multiproteicos/metabolismo , Estabilidad del ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...