Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Genet ; 13: 876862, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36092926

RESUMO

Chromatin is thought to regulate the accessibility of the underlying DNA sequence to machinery that transcribes and repairs the DNA. Heterochromatin is chromatin that maintains a sufficiently high density of DNA packing to be visible by light microscopy throughout the cell cycle and is thought to be most restrictive to transcription. Several studies have suggested that larger proteins and protein complexes are attenuated in their access to heterochromatin. In addition, heterochromatin domains may be associated with phase separated liquid condensates adding further complexity to the regulation of protein concentration within chromocenters. This provides a solvent environment distinct from the nucleoplasm, and proteins that are not size restricted in accessing this liquid environment may partition between the nucleoplasm and heterochromatin based on relative solubility. In this study, we assessed the accessibility of constitutive heterochromatin in mouse cells, which is organized into large and easily identifiable chromocenters, to fluorescently tagged DNA damage response proteins. We find that proteins larger than the expected 10 nm size limit can access the interior of heterochromatin. We find that the sensor proteins Ku70 and PARP1 enrich in mouse chromocenters. At the same time, MRE11 shows variability within an asynchronous population that ranges from depleted to enriched but is primarily homogeneously distribution between chromocenters and the nucleoplasm. While larger downstream proteins such as ATM, BRCA1, and 53BP1 are commonly depleted in chromocenters, they show a wide range of concentrations, with none being depleted beyond approximately 75%. Contradicting exclusively size-dependent accessibility, many smaller proteins, including EGFP, are also depleted in chromocenters. Our results are consistent with minimal size-dependent selectivity but a distinct solvent environment explaining reduced concentrations of diffusing nucleoplasmic proteins within the volume of the chromocenter.

2.
Neurotherapeutics ; 14(2): 274-283, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28233220

RESUMO

Progress in the treatment of adult high-grade gliomas (HGG), including chemoradiation with concurrent and adjuvant temozolomide for glioblastoma, has not translated into significant therapeutic advances for pediatric HGG, where overall survival has plateaued at 15% to 20%, especially when considering specialized pediatric treatment in tertiary care centers, maximal safe neurosurgical resection, optimized delivery of involved field radiation, and improvements in supportive care. However, recent advances in our understanding of pediatric HGG, including the application of next-generation sequencing and DNA methylation profiling, have identified mutations in the histone variant H3.3 and canonical H3.1 genes, respectively. These mutations are relatively specific to neuroanatomic compartments (cortex, midline structures, thalamus, brainstem) and are often associated with other mutations, especially in specific growth factor receptor tyrosine kinases. Targeting epigenetic pathways affected by these histone mutations, alone or in combination with small molecule inhibitors of growth factor receptor signaling pathways, will inform new treatment strategies for pediatric HGG and should be incorporated into novel cooperative group clinical trial designs.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Epigênese Genética , Glioma/genética , Glioma/terapia , Histonas/metabolismo , Adolescente , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Criança , Pré-Escolar , Metilação de DNA , Glioma/metabolismo , Glioma/patologia , Humanos , Mutação , Gradação de Tumores
3.
J Biol Chem ; 291(4): 1789-1802, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26559976

RESUMO

Chromatin undergoes a rapid ATP-dependent, ATM and H2AX-independent decondensation when DNA damage is introduced by laser microirradiation. Although the detailed mechanism of this decondensation remains to be determined, the kinetics of decondensation are similar to the kinetics of poly(ADP-ribosyl)ation. We used laser microirradiation to introduce DNA strand breaks into living cells expressing a photoactivatable GFP-tagged histone H2B. We find that poly(ADP-ribosyl)ation mediated primarily by poly(ADP-ribose) polymerase 1 (PARP1) is responsible for the rapid decondensation of chromatin at sites of DNA damage. This decondensation of chromatin correlates temporally with the displacement of histones, which is sensitive to PARP inhibition and is transient in nature. Contrary to the predictions of the histone shuttle hypothesis, we did not find that histone H1 accumulated on poly(ADP-ribose) (PAR) in vivo. Rather, histone H1, and to a lessor extent, histones H2A and H2B were rapidly depleted from the sites of PAR accumulation. However, histone H1 returns to chromatin and the chromatin recondenses. Thus, the PARP-dependent relaxation of chromatin closely correlates with histone displacement.


Assuntos
Montagem e Desmontagem da Cromatina/efeitos da radiação , Cromatina/metabolismo , Cromatina/efeitos da radiação , Histonas/metabolismo , Animais , Linhagem Celular , Dano ao DNA/efeitos da radiação , Reparo do DNA , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Humanos , Lasers , Camundongos , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo
4.
Anticancer Res ; 34(9): 4717-22, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25202049

RESUMO

BACKGROUND: Proper re-establishment of heterochromatin after each round of DNA replication is critical to the preservation of cell identity. Paired box 3 (PAX3), a transcription factor important in embryonic development, was found to mediate the formation of pericentromeric heterochromatin. However, how PAX3 recognizes the heterochromatic environment and re-establishes it after DNA replication remains unclear. MATERIALS AND METHODS: Cell-cycle synchronization, fluorescence microscopic analyses, and co-immunoprecipitation were used to analyze the heterochromatic localization of PAX3 in HEK 293 cells and NIH 3T3 cells. RESULTS: We found that PAX3 binds pericentromeric heterochromatin during middle-to-late S phase. Loading of PAX3 onto pericentromeric heterochromatin requires poly(ADP-ribose) polymerase 1 (PARP1). Furthermore, loss of PAX3 or PARP1 delays cell-cycle progression through the S phase. CONCLUSION: Our results reveal how PAX3 recognizes and maintains pericentromeric heterochromatin at the S phase of the cell cycle.


Assuntos
Heterocromatina/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Fase S , Animais , Ciclo Celular , Células HEK293 , Heterocromatina/genética , Humanos , Camundongos , Células NIH 3T3 , Fatores de Transcrição Box Pareados/genética , Ligação Proteica , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo
5.
J Mol Biol ; 402(1): 178-93, 2010 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-20643146

RESUMO

The transcription factor PAX3 is essential for myogenesis and neural crest development, and is one of several genes mutated in human Waardenburg syndrome. Analysis of disease-causing missense mutations in PAX3 has established the interdependence of its two DNA-binding domains, the paired domain (PD) and the homeodomain (HD), as well as defects in localization and mobility. Paradoxically, mutants that retained DNA binding activity exhibited the greatest defects in localization and mobility, regardless of the domain in which they reside. In the present study, structure-function analyses were used to determine the mechanistic basis of this effect. In the context of the isolated DNA-binding domains, HD mutants adopted an increase in mobility proportional to their loss in DNA binding, while PD mutants continued to display the inverse relationship observed in the full-length protein. At the structural level, this reflected an unexpected dependence on base-specific contacts in the PD, whereas HD mobility was more severely affected by loss of backbone contacts, as has been observed with other DNA-binding proteins. This requires that the HD switch to a base-specific mode in the full-length protein. Moreover, both domains underwent substantial reduction in mobility and altered localization when in a contiguous polypeptide with the endogenous linker segment. Notably, although the HD conferred localization to heterochromatin, this activity was masked when linked to the PD, despite the absence of determinants for subnuclear compartmentalization in the PD or linker. Last, the propensity for PAX3 heterochromatin localization was modulated by sequences at the amino and carboxy termini, supporting a model in which alternate conformations lead to unmasking of the HD. These data indicate that the PD and the HD functionally interact in vivo and behave as a single binding module whose mobility and localization are dependent on sequence-specific contacts.


Assuntos
DNA/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Animais , Núcleo Celular/metabolismo , Células Cultivadas , DNA/genética , Embrião de Mamíferos , Fibroblastos , Proteínas de Homeodomínio , Humanos , Camundongos , Mutação/genética , Sinais de Localização Nuclear , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/química , Fatores de Transcrição Box Pareados/genética , Estrutura Terciária de Proteína , Frações Subcelulares
6.
J Biol Chem ; 281(13): 8888-97, 2006 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-16373353

RESUMO

Histone methylation is unique among post-translational histone modifications by virtue of its stability. It is thought to be a relatively stable and heritable epigenetic mark for gene-specific regulation. In this study, we use quantitative in situ approaches to investigate the cell cycle dynamics of methylated isoforms of histone H3 lysine 9. Contrary to the expected stability of trimethylated lysines, our results for trimethylated lysine 9 (tMeK9) of H3 demonstrate that the genomic content of this methylation undergoes significant changes as cells progress through mitosis. Unexpectedly, there is a loss of tMeK9 that appears to reflect a robust demethylase activity that is active during the period between anaphase and cytokinesis. Subsequent investigations of mitoses in tMeK9-deficient cells revealed defects in chromosome congression and segregation that are distinct from the increased cohesion at centromeres previously reported in association with the loss of tMeK9. Collectively, these results identify a mitosis-specific trimethylation of Lys9 in pericentromeric heterochromatin that functions in the faithful segregation of chromosomes.


Assuntos
Segregação de Cromossomos , Cromossomos Humanos , Histonas/metabolismo , Lisina/análogos & derivados , Lisina/química , Mitose , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular , Linhagem Celular Transformada , Inibidores Enzimáticos/farmacologia , Fibroblastos/metabolismo , Citometria de Fluxo , Técnica Direta de Fluorescência para Anticorpo , Corantes Fluorescentes , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Histonas/genética , Humanos , Immunoblotting , Indóis , Metilação , Cervo Muntjac , Nocodazol/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Pele/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA