Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Differ ; 27(1): 29-43, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31065107

RESUMO

Somatic oncogenic mutation of BRAF coupled with inactivation of PTEN constitute a frequent combination of genomic alterations driving the development of human melanoma. Mice genetically engineered to conditionally express oncogenic BrafV600E and inactivate Pten in melanocytes following tamoxifen treatment rapidly develop melanoma. While early-stage melanomas comprised melanin-pigmented Mitf and Dct-expressing cells, expression of these and other melanocyte identity genes was lost in later stage tumours that showed histological and molecular characteristics of de-differentiated neural crest type cells. Melanocyte identity genes displayed loss of active chromatin marks and RNA polymerase II and gain of heterochromatin marks, indicating epigenetic reprogramming during tumour progression. Nevertheless, late-stage tumour cells grown in culture re-expressed Mitf, and melanocyte markers and Mitf together with Sox10 coregulated a large number of genes essential for their growth. In this melanoma model, somatic inactivation that the catalytic Brg1 (Smarca4) subunit of the SWI/SNF complex and the scaffolding Bptf subunit of the NuRF complex delayed tumour formation and deregulated large and overlapping gene expression programs essential for normal tumour cell growth. Moreover, we show that Brg1 and Bptf coregulated many genes together with Mitf and Sox10. Together these transcription factors and chromatin remodelling complexes orchestrate essential gene expression programs in mouse melanoma cells.


Assuntos
Antígenos Nucleares/fisiologia , DNA Helicases/fisiologia , Regulação Neoplásica da Expressão Gênica , Melanoma Experimental/genética , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Cutâneas/genética , Fatores de Transcrição/fisiologia , Animais , Antígenos Nucleares/genética , DNA Helicases/genética , Progressão da Doença , Epigênese Genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , PTEN Fosfo-Hidrolase/genética , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Fatores de Transcrição/genética , Células Tumorais Cultivadas
2.
Nucleic Acids Res ; 47(6): 2703-2715, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30812030

RESUMO

P53-binding protein 1 (53BP1) mediates DNA repair pathway choice and promotes checkpoint activation. Chromatin marks induced by DNA double-strand breaks and recognized by 53BP1 enable focal accumulation of this multifunctional repair factor at damaged chromatin. Here, we unveil an additional level of regulation of 53BP1 outside repair foci. 53BP1 movements are constrained throughout the nucleoplasm and increase in response to DNA damage. 53BP1 interacts with the structural protein NuMA, which controls 53BP1 diffusion. This interaction, and colocalization between the two proteins in vitro and in breast tissues, is reduced after DNA damage. In cell lines and breast carcinoma NuMA prevents 53BP1 accumulation at DNA breaks, and high NuMA expression predicts better patient outcomes. Manipulating NuMA expression alters PARP inhibitor sensitivity of BRCA1-null cells, end-joining activity, and immunoglobulin class switching that rely on 53BP1. We propose a mechanism involving the sequestration of 53BP1 by NuMA in the absence of DNA damage. Such a mechanism may have evolved to disable repair functions and may be a decisive factor for tumor responses to genotoxic treatments.


Assuntos
Antígenos Nucleares/fisiologia , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Proteínas Associadas à Matriz Nuclear/fisiologia , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Reparo do DNA por Junção de Extremidades/genética , Regulação para Baixo , Feminino , Células HEK293 , Humanos , Ligação Proteica
3.
Cancer Res ; 76(21): 6183-6192, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27651309

RESUMO

Genetic studies in fruit flies have implicated the chromatin remodeling complex nucleosome remodeling factor (NURF) in immunity, but it has yet to be studied in mammals. Here we show that its targeting in mice enhances antitumor immunity in two syngeneic models of cancer. NURF was disabled by silencing of bromodomain PHD-finger containing transcription factor (BPTF), the largest and essential subunit of NURF. We found that both CD8+ and CD4+ T cells were necessary for enhanced antitumor activity, with elevated numbers of activated CD8+ T cells observed in BPTF-deficient tumors. Enhanced cytolytic activity was observed for CD8+ T cells cocultured with BPTF-silenced cells. Similar effects were not produced with T-cell receptor transgenic CD8+ T cells, implicating the involvement of novel antigens. Accordingly, enhanced activity was observed for individual CD8+ T-cell clones from mice bearing BPTF-silenced tumors. Mechanistic investigations revealed that NURF directly regulated the expression of genes encoding immunoproteasome subunits Psmb8 and Psmb9 and the antigen transporter genes Tap1 and Tap2 The PSMB8 inhibitor ONX-0914 reversed the effects of BPTF ablation, consistent with a critical role for the immunoproteasome in improving tumor immunogenicity. Thus, NURF normally suppresses tumor antigenicity and its depletion improves antigen processing, CD8 T-cell cytotoxicity, and antitumor immunity, identifying NURF as a candidate therapeutic target to enhance antitumor immunity. Cancer Res; 76(21); 6183-92. ©2016 AACR.


Assuntos
Antígenos Nucleares/fisiologia , Neoplasias/imunologia , Proteínas do Tecido Nervoso/fisiologia , Linfócitos T/imunologia , Fatores de Transcrição/fisiologia , Animais , Apresentação de Antígeno , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Nucleossomos/fisiologia
4.
Sci Rep ; 6: 23588, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-27026195

RESUMO

Neuropilins (NRPs) are cell surface glycoproteins that often act as co-receptors for plexins and VEGF family receptors. Neuropilin-2 (NRP2), a family member of NRPs, was shown to regulate autophagy and endocytic trafficking in cancer cells, a function distinctly different from its role as a co-receptor. WD Repeat and FYVE domain containing 1 (WDFY1)-protein acts downstream of NRP2 for this function. Our results indicated that NRP2 maintains an optimum concentration of WDFY1 by negatively regulating its expression. Since increased expression of WDFY1 reduces the endocytic activity, maintenance of WDFY1 level is crucial in metastatic cancer cells to sustain high endocytic activity, essential for promotion of oncogenic activation and cancer cell survival. Here, we have delineated the underlying molecular mechanism of WDFY1 synthesis by NRP2. Our results indicated that NRP2 inhibits WDFY1 transcription by preventing the nuclear localization of a transcription factor, Fetal ALZ50-reactive clone 1 (FAC1). Our finding is novel as transcriptional regulation of a gene by NRP2 axis has not been reported previously. Regulation of WDFY1 transcription by NRP2 axis is a critical event in maintaining metastatic phenotype in cancer cells. Thus, inhibiting NRP2 or hyper-activating WDFY1 can be an effective strategy to induce cell death in metastatic cancer.


Assuntos
Antígenos Nucleares/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neuropilina-2/fisiologia , Proteínas Nucleares/genética , Fatores de Transcrição/fisiologia , Ativação Transcricional , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Humanos , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Estabilidade Proteica , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica
5.
Mol Neurobiol ; 53(3): 1637-1647, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25680637

RESUMO

Neuronal nuclei (NeuN) is a well-recognized "marker" that is detected exclusively in post-mitotic neurons and was initially identified through an immunological screen to produce neuron-specific antibodies. Immunostaining evidence indicates that NeuN is distributed in the nuclei of mature neurons in nearly all parts of the vertebrate nervous system. NeuN is highly conserved among species and is stably expressed during specific stages of development. Therefore, NeuN has been considered to be a reliable marker of mature neurons for the past two decades. However, this role has been challenged by recent studies indicating that NeuN staining is variable and even absent during certain diseases and specific physiological states. More importantly, despite the widespread use of the anti-NeuN antibody, the natural identity of the NeuN protein remained elusive for 17 years. NeuN was recently eventually identified as an epitope of Rbfox3, which is a novel member of the Rbfox1 family of splicing factors. This identification might provide a novel perspective on NeuN expression during both physiological and pathological conditions. This review summarizes the current progress on the biochemical identity and biological significance of NeuN and recommends caution when applying NeuN immunoreactivity as a definitive marker of mature neurons in certain diseases and specific physiological states.


Assuntos
Processamento Alternativo , Antígenos Nucleares/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Fatores de Processamento de RNA/fisiologia , Animais , Axotomia , Biomarcadores , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Epitopos , Regulação da Expressão Gênica , Humanos , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/metabolismo , Neurônios/química , Estresse Fisiológico , Vertebrados/metabolismo
6.
Front Biosci (Landmark Ed) ; 21(3): 514-27, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26709791

RESUMO

Non-homologous end-joining (NHEJ) is an essential DNA double strand break repair pathway during all cell cycle stages. Deficiency in NHEJ factors can lead to accumulation of unrepaired DNA breaks or faulty DNA repair, which may ultimately result in cell death, senescence or carcinogenesis. The Ku70/80 heterodimer is a key-player in the NHEJ pathway and binds to DNA termini with high affinity, where it helps to protect DNA ends from degradation and to recruit other NHEJ factors required for repair. The mechanism of Ku70/80 detachment from the DNA helix after completion of DNA repair is incompletely understood. Some data suggest that certain DNA repair factors are ubiquitylated and targeted for proteasomal degradation after repair. Recent studies suggest that Ku80 is conjugated to lysine48-linked ubiquitin chains by the Skp1-Cullin-F-box (SCF) complex and/or the RING finger protein 8 (RNF8) ubiquitin-protein ligases, followed by rapid proteasomal degradation. In this review we address the structure and function of the Ku70/80 heterodimer and how ubiquitylation may affect the release of Ku70/80 from chromatin and its subsequent degradation via the ubiquitin-proteasome system.


Assuntos
Antígenos Nucleares/fisiologia , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/fisiologia , Dano ao DNA , Reparo do DNA , Autoantígeno Ku , Ubiquitina-Proteína Ligases/metabolismo
7.
Mol Cell ; 58(5): 743-54, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-25936801

RESUMO

The circadian clock orchestrates global changes in transcriptional regulation on a daily basis via the bHLH-PAS transcription factor CLOCK:BMAL1. Pathways driven by other bHLH-PAS transcription factors have a homologous repressor that modulates activity on a tissue-specific basis, but none have been identified for CLOCK:BMAL1. We show here that the cancer/testis antigen PASD1 fulfills this role to suppress circadian rhythms. PASD1 is evolutionarily related to CLOCK and interacts with the CLOCK:BMAL1 complex to repress transcriptional activation. Expression of PASD1 is restricted to germline tissues in healthy individuals but can be induced in cells of somatic origin upon oncogenic transformation. Reducing PASD1 in human cancer cells significantly increases the amplitude of transcriptional oscillations to generate more robust circadian rhythms. Our results describe a function for a germline-specific protein in regulation of the circadian clock and provide a molecular link from oncogenic transformation to suppression of circadian rhythms.


Assuntos
Antígenos de Neoplasias/fisiologia , Antígenos Nucleares/fisiologia , Proteínas CLOCK/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Sequência de Aminoácidos , Antígenos de Neoplasias/química , Antígenos Nucleares/química , Proteínas CLOCK/metabolismo , Linhagem Celular Tumoral , Ritmo Circadiano , Sequência Conservada , Éxons , Humanos , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Testículo/metabolismo
8.
J Vet Med Sci ; 77(9): 1137-42, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25947323

RESUMO

To elucidate the mechanisms of DNA repair pathway is critical for developing next-generation radiotherapies and chemotherapeutic drugs for cancer. Ionizing radiation and many chemotherapeutic drugs kill tumor cells mainly by inducing DNA double-strand breaks (DSBs). The classical nonhomologous DNA-end joining (NHEJ) (C-NHEJ) pathway repairs a predominant fraction of DSBs in mammalian cells. The C-NHEJ pathway appears to start with the binding of Ku (heterodimer of Ku70 and Ku80) to DNA break ends. Therefore, recruitment of Ku to DSB sites might play a critical role in regulating NHEJ activity. Indeed, human Ku70 and Ku80 localize in the nuclei and accumulate at microirradiated DSB sites. However, the localization and regulation mechanisms of Ku70 and Ku80 homologues in animal models, such as mice and other species, have not been elucidated in detail, particularly in cells immediately after microirradiation. Here, we show that EYFP-tagged mouse Ku70 localizes in the interphase nuclei of mouse fibroblasts and epithelial cells. Furthermore, our findings indicate that EYFP-mouse Ku70 accumulates with its heterodimeric partner Ku80 immediately at laser-microirradiated DSB sites. We also confirmed that the structure of Ku70 nuclear localization signal (NLS) is highly conserved among various rodent species, such as the mouse, rat, degu and ground squirrel, supporting the idea that NLS is important for the regulation of rodent Ku70 function. Collectively, these results suggest that the mechanisms of regulating the localization and accumulation of Ku70 at DSBs might be well conserved between the mouse and human species.


Assuntos
Antígenos Nucleares/efeitos da radiação , Núcleo Celular/metabolismo , Dano ao DNA/efeitos da radiação , Proteínas de Ligação a DNA/efeitos da radiação , Interfase/efeitos da radiação , Animais , Antígenos Nucleares/fisiologia , Linhagem Celular , Núcleo Celular/efeitos da radiação , Reparo do DNA por Junção de Extremidades/efeitos da radiação , Reparo do DNA/efeitos da radiação , Proteínas de Ligação a DNA/fisiologia , Imunofluorescência , Autoantígeno Ku , Camundongos
9.
Nucleic Acids Res ; 43(4): 2138-51, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25653166

RESUMO

The Ku heterodimer serves in the initial step in repairing DNA double-strand breaks by the non-homologous end-joining pathway. Besides this key function, Ku also plays a role in other cellular processes including telomere maintenance. Inactivation of Ku can lead to DNA repair defects and telomere aberrations. In model organisms where Ku has been studied, inactivation can lead to DNA repair defects and telomere aberrations. In general Ku deficient mutants are viable, but a notable exception to this is human where Ku has been found to be essential. Here we report that similar to the situation in human Ku is required for cell proliferation in the fungus Ustilago maydis. Using conditional strains for Ku expression, we found that cells arrest permanently in G2 phase when Ku expression is turned off. Arrest results from cell cycle checkpoint activation due to persistent signaling via the DNA damage response (DDR). Our results point to the telomeres as the most likely source of the DNA damage signal. Inactivation of the DDR makes the Ku complex dispensable for proliferation in this organism. Our findings suggest that in U. maydis, unprotected telomeres arising from Ku depletion are the source of the signal that activates the DDR leading to cell cycle arrest.


Assuntos
Antígenos Nucleares/fisiologia , Reparo do DNA , Proteínas de Ligação a DNA/fisiologia , Proteínas Fúngicas/fisiologia , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Telômero/metabolismo , Antígenos Nucleares/genética , Dano ao DNA , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Autoantígeno Ku , Transdução de Sinais , Telômero/química , Homeostase do Telômero , Ustilago/genética
10.
Int J Cancer ; 136(6): 1263-75, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25082302

RESUMO

Squamous cell carcinoma (SCC) occurring in the head and neck region and the esophagus causes tremendous cancer mortality around the world. miR-31 is among the most eminently upregulated MicroRNAs in SCC, when it occurs in the head and neck region and the esophagus. We established miR-31 transgenic mouse lines, in which miR-31 is under the control of the K14 promoter. 4-nitroquinoline 1-oxide (4NQO) is a mutagen that causes double strand breaks. The transgenic mice exhibited a higher potential for tumor induction than wild-type (Wt) mice of the tongue and esophagus after 4NQO treatment. After 4NQO treatment or irradiation, p-γH2AX expression in squamous epithelium of transgenic mice was increased more than in Wt mice. Exogenous expression of miR-31 was also found to be associated with the higher p-γH2AX expression induced by 4NQO in human oral SCC (OSCC) cell lines. The repair genes PARP1 and Ku80 were validated as new targets of miR-31 in human OSCC cell lines, and were found to be downregulated in the squamous epithelium of the tongue in transgenic mice. However, only the downregulation of Ku80 was essential for maintaining the high level of p-γH2AX induced by 4NQO in OSCC cells. Inverse expression profiles for miR-31 and Ku80 were noted in human OSCC tissue. Our study identifies the high sensitivity of K14-EGFP-miR-31 transgenic mice to chemical carcinogen-induced squamous cell tumorigenesis and shows that this seems to be associated with the downregulation of Ku80 and an impairment of repair activity in squamous cells, which are mediated by miR-31.


Assuntos
4-Nitroquinolina-1-Óxido/toxicidade , Antígenos Nucleares/fisiologia , Carcinoma de Células Escamosas/induzido quimicamente , Proteínas de Ligação a DNA/fisiologia , MicroRNAs/fisiologia , Neoplasias Bucais/induzido quimicamente , Animais , Linhagem Celular Tumoral , Dano ao DNA , Neoplasias Esofágicas/induzido quimicamente , Proteínas de Fluorescência Verde/metabolismo , Histonas/análise , Humanos , Autoantígeno Ku , Camundongos , Camundongos Transgênicos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/fisiologia
11.
Semin Cell Dev Biol ; 34: 140-5, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24972323

RESUMO

The angle of cell division is critical in at least two contexts. It can determine cell fate, as it does in developing neural tissue. It can also dictate tissue architecture, as it does in many epithelia. One way to ensure the correct angle of cell division is through controlled orientation of the spindle at metaphase. What happens when that control is lost? Ongoing work suggests that the consequence of metaphase spindle misorientation may be significant, but multiple mechanisms exist to protect the cell and the tissue. We speculate that one such mechanism involves a recently identified anaphase activity for two of the key players at metaphase: NuMA (Mud, LIN-5) and dynein.


Assuntos
Fuso Acromático/metabolismo , Animais , Antígenos Nucleares/fisiologia , Carcinogênese/patologia , Proteínas de Ciclo Celular , Sistema Nervoso Central/patologia , Humanos , Mitose , Neoplasias/patologia , Proteínas Associadas à Matriz Nuclear/fisiologia , Fuso Acromático/patologia
12.
PLoS One ; 9(4): e93568, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24740260

RESUMO

Ku80 and DNA-PKCS are both involved in the repair of double strand DNA breaks via the nonhomologous end joining (NHEJ) pathway. While ku80-/- mice exhibit a severely reduced lifespan and size, this phenotype is less pronounced in dna-pkcs-/- mice. However, these observations are based on independent studies with varying genetic backgrounds. Here, we generated ku80-/-, dna-pkcs-/- and double knock out mice in a C57Bl6/J*FVB F1 hybrid background and compared their lifespan, end of life pathology and mutation frequency in liver and spleen using a lacZ reporter. Our data confirm that inactivation of Ku80 and DNA-PKCS causes reduced lifespan and bodyweights, which is most severe in ku80-/- mice. All mutant mice exhibited a strong increase in lymphoma incidence as well as other aging-related pathology (skin epidermal and adnexal atrophy, trabacular bone reduction, kidney tubular anisokaryosis, and cortical and medullar atrophy) and severe lymphoid depletion. LacZ mutation frequency analysis did not show strong differences in mutation frequencies between knock out and wild type mice. The ku80-/- mice had the most severe phenotype and the Ku80-mutation was dominant over the DNA-PKCS-mutation. Presumably, the more severe degenerative effect of Ku80 inactivation on lifespan compared to DNA-PKCS inactivation is caused by additional functions of Ku80 or activity of free Ku70 since both Ku80 and DNA-PKCS are essential for NHEJ.


Assuntos
Envelhecimento/genética , Antígenos Nucleares/genética , Reparo do DNA por Junção de Extremidades/genética , Proteína Quinase Ativada por DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas Nucleares/genética , Animais , Antígenos Nucleares/fisiologia , Peso Corporal/genética , Dano ao DNA , Proteína Quinase Ativada por DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Feminino , Técnicas de Inativação de Genes , Autoantígeno Ku , Longevidade/genética , Masculino , Camundongos Endogâmicos C57BL , Proteínas Nucleares/fisiologia , Fenótipo
13.
Cell Death Differ ; 21(7): 1160-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24769731

RESUMO

Mcl-1 is a unique antiapoptotic Bcl2 family member with a short half-life due to its rapid turnover through ubiquitination. We discovered that Ku70, a DNA double-strand break repair protein, functions as a deubiquitinase to stabilize Mcl-1. Ku70 knockout in mouse embryonic fibroblast (MEF) cells or depletion from human lung cancer H1299 cells leads to the accumulation of polyubiquitinated Mcl-1 and a reduction in its half-life and protein expression. Conversely, expression of exogenous Ku70 in Ku70(-/-) MEF cells restores Mcl-1 expression. Subcellular fractionation indicates that Ku70 extensively colocalizes with Mcl-1 in mitochondria, endoplasmic reticulum and nucleus in H1299 cells. Ku70 directly interacts with Mcl-1 via its C terminus (that is, aa 536-609), which is required and sufficient for deubiquitination and stabilization of Mcl-1, leading to suppression of apoptosis. Purified Ku70 protein directly deubiquitinates Mcl-1 by removing K48-linked polyubiquitin chains. Ku70 knockdown not only promotes Mcl-1 turnover but also enhances antitumor efficacy of the BH3-mimetic ABT-737 in human lung cancer xenografts. These findings identify Ku70 as a novel Mcl-1 deubiquitinase that could be a potential target for cancer therapy by manipulating Mcl-1 deubiquitination.


Assuntos
Antígenos Nucleares/fisiologia , Apoptose , Proteínas de Ligação a DNA/fisiologia , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Ubiquitinação , Animais , Antineoplásicos/farmacologia , Compostos de Bifenilo/farmacologia , Sobrevivência Celular , Células HCT116 , Células HEK293 , Meia-Vida , Humanos , Autoantígeno Ku , Camundongos , Camundongos Nus , Nitrofenóis/farmacologia , Piperazinas/farmacologia , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Transporte Proteico , Estaurosporina/farmacologia , Sulfonamidas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Nucleic Acids Res ; 42(10): 6365-79, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24753406

RESUMO

Chromatin remodeling factors play an active role in the DNA damage response by shaping chromatin to facilitate the repair process. The spatiotemporal regulation of these factors is key to their function, yet poorly understood. We report that the structural nuclear protein NuMA accumulates at sites of DNA damage in a poly[ADP-ribose]ylation-dependent manner and functionally interacts with the ISWI ATPase SNF2h/SMARCA5, a chromatin remodeler that facilitates DNA repair. NuMA coimmunoprecipitates with SNF2h, regulates its diffusion in the nucleoplasm and controls its accumulation at DNA breaks. Consistent with NuMA enabling SNF2h function, cells with silenced NuMA exhibit reduced chromatin decompaction after DNA cleavage, lesser focal recruitment of homologous recombination repair factors, impaired DNA double-strand break repair in chromosomal (but not in episomal) contexts and increased sensitivity to DNA cross-linking agents. These findings reveal a structural basis for the orchestration of chromatin remodeling whereby a scaffold protein promotes genome maintenance by directing a remodeler to DNA breaks.


Assuntos
Adenosina Trifosfatases/metabolismo , Antígenos Nucleares/fisiologia , Proteínas Cromossômicas não Histona/metabolismo , Quebras de DNA de Cadeia Dupla , Proteínas Associadas à Matriz Nuclear/fisiologia , Reparo de DNA por Recombinação , Proteínas de Ciclo Celular , Linhagem Celular , Linhagem Celular Tumoral , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Histonas/metabolismo , Humanos
15.
Oncogene ; 33(21): 2748-57, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-23752193

RESUMO

Ku70, a known nonhomologous end-joining (NHEJ) factor, also functions in tumor suppression, although this molecular mechanism remains uncharacterized. Previously, we showed that mice deficient for DNA ligase IV (Lig4), another key NHEJ factor, succumbed to aggressive lymphoma in the absence of tumor suppressor p53. However, the tumor phenotype is abrogated by the introduction of a hypomorphic mutant p53(R172P), which impaired p53-mediated apoptosis but not cell-cycle arrest. However, Lig4(-/-)p53(R172P) mice succumbed to severe diabetes. To further elucidate the role of NHEJ and p53-mediated apoptosis in vivo, we bred Ku70(-/-) p53(R172P) mice. Unexpectedly, these mice were free of diabetes, although 80% of the mutant mice had abnormally enlarged colons with pronounced inflammation. Remarkably, most of these mutant mice progressed to dysplasia, adenoma and adenocarcinoma; this is in contrast to the Lig4(-/-)p53(R172P) phenotype, strongly suggesting an NHEJ-independent function of Ku70. Significantly, our analyses of Ku70(-/-)p53(R172P) colonic epithelial cells show nuclear stabilization of ß-catenin accompanied by higher expression of cyclin D1 and c-Myc in affected colon sections than in control samples. This is not due to the p53 mutation, as Ku70(-/-) mice share this phenotype. Our results not only unravel a novel function of Ku70 essential for colon homeostasis, but also establish an excellent in vivo model in which to study how chronic inflammation and abnormal cellular proliferation underlie tumorigenesis and tumor progression in the colon.


Assuntos
Adenocarcinoma/genética , Antígenos Nucleares/fisiologia , Neoplasias Colorretais/genética , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/fisiologia , Homeostase , Adenocarcinoma/metabolismo , Animais , Carcinogênese , Proliferação de Células , Células Cultivadas , Senescência Celular , Colo/imunologia , Colo/patologia , Neoplasias Colorretais/metabolismo , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Células Epiteliais/metabolismo , Humanos , Interleucina-6/metabolismo , Autoantígeno Ku , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-myc/metabolismo , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/genética , Via de Sinalização Wnt
16.
Cell Mol Life Sci ; 71(14): 2731-45, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24305947

RESUMO

DNA double-strand breaks (DSBs) can cause either cell death or genomic instability. The Ku heterodimer Ku70/80 is required for the NHEJ (non-homologous end-joining) DNA DSB repair pathway. The INHAT (inhibitor of histone acetyltransferases) complex subunit, SET/TAF-Iß, can inhibit p300- and PCAF-mediated acetylation of both histone and p53, thereby repressing general transcription and that of p53 target genes. Here, we show that SET/TAF-Iß interacts with Ku70/80, and that this interaction inhibits CBP- and PCAF-mediated Ku70 acetylation in an INHAT domain-dependent manner. Notably, DNA damage by UV disrupted the interaction between SET/TAF-Iß and Ku70. Furthermore, we demonstrate that overexpressed SET/TAF-Iß inhibits recruitment of Ku70/80 to DNA damage sites. We propose that dysregulation of SET/TAF-Iß expression prevents repair of damaged DNA and also contributes to cellular proliferation. All together, our findings indicate that SET/TAF-Iß interacts with Ku70/80 in the nucleus and inhibits Ku70 acetylation. Upon DNA damage, SET/TAF-Iß dissociates from the Ku complex and releases Ku70/Ku80, which are then recruited to DNA DSB sites via the NHEJ DNA repair pathway.


Assuntos
Antígenos Nucleares/fisiologia , Dano ao DNA , Reparo do DNA por Junção de Extremidades/fisiologia , Proteínas de Ligação a DNA/fisiologia , Chaperonas de Histonas/fisiologia , Fatores de Transcrição/fisiologia , Acetilação , Antígenos Nucleares/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Chaperonas de Histonas/metabolismo , Humanos , Autoantígeno Ku , Modelos Genéticos , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Fatores de Transcrição/metabolismo
17.
Igaku Butsuri ; 34(2): 57-64, 2014.
Artigo em Japonês | MEDLINE | ID: mdl-25693292

RESUMO

DNA double-strand break (DSB) is considered most deleterious among radiation-induced DNA damages and most relevant to the biological effects of radiation. In eukaryotic cells, DSB is repaired mainly through two pathways: non-homologous end-joining (NHEJ) and homologous recombination (HR). These repair pathways seem to play complementary roles. NHEJ is considered less accurate than HR, but HR is available only in late S and G2 phases in vertebrates. Recent studies elucidated how cells choose one from these two pathways depending on the circumstance: cell cycle phase, complexity of DNA damage and chromatin structure.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA por Junção de Extremidades/genética , Reparo do DNA por Junção de Extremidades/fisiologia , Reparo de DNA por Recombinação/genética , Reparo de DNA por Recombinação/fisiologia , Transportadores de Cassetes de Ligação de ATP/fisiologia , Hidrolases Anidrido Ácido , Animais , Antígenos Nucleares/fisiologia , Proteínas de Transporte/fisiologia , Proteínas de Ciclo Celular/fisiologia , Cromatina , Enzimas Reparadoras do DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Fase G2/genética , Fase G2/fisiologia , Humanos , Autoantígeno Ku , Proteína Homóloga a MRE11 , Camundongos , Proteínas Nucleares/fisiologia , Radiobiologia , Fase S/genética , Fase S/fisiologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/fisiologia
18.
Radiother Oncol ; 108(3): 429-33, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23932191

RESUMO

BACKGROUND AND PURPOSE: In muscle-invasive bladder cancer there is an urgent need to identify relatively non-toxic radiosensitising agents for use in elderly patients. Histone deacetylase inhibitors radiosensitise tumour cells but not normal cells in vitro and variously downregulate DNA damage signalling, homologous recombination (HR) and non-homologous end-joining (NHEJ) repair proteins. We investigated panobinostat (PAN) as a potential radiosensitiser in bladder cancer cells. MATERIALS AND METHODS: Clonogenic assays were performed in RT112 bladder cancer cells, and RT112 cells stably knocked down for RAD51 or Ku80 by shRNAi. Resolution of γH2AX foci was determined by immunofluorescence confocal microscopy, cell cycle progression by FACS analysis and protein expression by western blotting. RESULTS: PAN had a greater radiosensitising effect in Ku80KD than RT112 or RAD51KD cells; enhancement ratios 1.35 for Ku80KD at 10nM (IC(20) for Ku80KD) and 1.31 for RT112 and RAD51KD at 25 nM (IC(40) for both). PAN downregulated MRE11, NBS1 and RAD51, but not Ku70 and Ku80, increased γH2AX foci formation in a dose-dependent manner and delayed γH2AX foci repair after ionising radiation. CONCLUSIONS: PAN acts as a radiosensitiser in bladder cancer cell lines, and appears to target HR rather than NHEJ. As muscle-invasive bladder tumours have reduced Ku-DNA binding, PAN could be particularly useful as a radiosensitiser in bladder cancer.


Assuntos
Antígenos Nucleares/fisiologia , Proteínas de Ligação a DNA/fisiologia , Ácidos Hidroxâmicos/farmacologia , Indóis/farmacologia , Radiossensibilizantes/farmacologia , Neoplasias da Bexiga Urinária/radioterapia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/análise , Histonas/análise , Humanos , Autoantígeno Ku , Proteína Homóloga a MRE11 , Panobinostat , Rad51 Recombinase/análise , Neoplasias da Bexiga Urinária/patologia
20.
Int J Radiat Oncol Biol Phys ; 86(3): 440-9, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23433795

RESUMO

DNA damage can occur as a result of endogenous metabolic reactions and replication stress or from exogenous sources such as radiation therapy and chemotherapy. DNA double strand breaks are the most cytotoxic form of DNA damage, and defects in their repair can result in genome instability, a hallmark of cancer. The major pathway for the repair of ionizing radiation-induced DSBs in human cells is nonhomologous end joining. Here we review recent advances on the mechanism of nonhomologous end joining, as well as new findings on its component proteins and regulation.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/fisiologia , Antígenos Nucleares/fisiologia , DNA Ligase Dependente de ATP , DNA Ligases/fisiologia , Enzimas Reparadoras do DNA/fisiologia , Proteína Quinase Ativada por DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Humanos , Autoantígeno Ku , Proteínas Nucleares/fisiologia , Fosforilação/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...