Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hum Mol Genet ; 28(12): 1947-1958, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30690483

RESUMO

UBE3A is a gene responsible for the pathogenesis of Angelman syndrome (AS), a neurodevelopmental disorder characterized by symptoms such as intellectual disability, delayed development and severe speech impairment. UBE3A encodes an E3 ubiquitin ligase, for which several targets have been identified, including synaptic molecules. Although proteolysis mainly occurs in the cytoplasm, UBE3A is localized to the cytoplasm and the nucleus. In fact, UBE3A is also known as a transcriptional regulator of the family of nuclear receptors. However, the function of UBE3A in the nucleus remains unclear. Therefore, we examined the involvement of UBE3A in transcription in the nuclei of neurons. Genome-wide transcriptome analysis revealed an enrichment of genes downstream of interferon regulatory factor (IRF) in a UBE3A-deficient AS mouse model. In vitro biochemical analyses further demonstrated that UBE3A interacted with IRF and, more importantly, that UBE3A enhanced IRF-dependent transcription. These results suggest a function for UBE3A as a transcriptional regulator of the immune system in the brain. These findings also provide informative molecular insights into the function of UBE3A in the brain and in AS pathogenesis.


Assuntos
Síndrome de Angelman/genética , Encéfalo/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Neurônios/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Síndrome de Angelman/metabolismo , Animais , Antivirais/metabolismo , Encéfalo/imunologia , Núcleo Celular/enzimologia , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células HEK293 , Humanos , Imunidade , Deficiência Intelectual/genética , Fator Regulador 2 de Interferon/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/enzimologia , Transcriptoma , Ubiquitina-Proteína Ligases/genética
2.
Cell Rep ; 10(8): 1310-23, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25732822

RESUMO

The 5S ribonucleoprotein particle (RNP) complex, consisting of RPL11, RPL5, and 5S rRNA, is implicated in p53 regulation under ribotoxic stress. Here, we show that the 5S RNP contributes to p53 activation and promotes cellular senescence in response to oncogenic or replicative stress. Oncogenic stress accelerates rRNA transcription and replicative stress delays rRNA processing, resulting in RPL11 and RPL5 accumulation in the ribosome-free fraction, where they bind MDM2. Experimental upregulation of rRNA transcription or downregulation of rRNA processing, mimicking the nucleolus under oncogenic or replicative stress, respectively, also induces RPL11-mediated p53 activation and cellular senescence. We demonstrate that exogenous expression of certain rRNA-processing factors rescues the processing defect, attenuates p53 accumulation, and increases replicative lifespan. To summarize, the nucleolar-5S RNP-p53 pathway functions as a senescence inducer in response to oncogenic and replicative stresses.


Assuntos
Senescência Celular , Ribossomos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Nucléolo Celular/metabolismo , Células Cultivadas , Humanos , Células MCF-7 , Camundongos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Interferência de RNA , RNA Ribossômico 5S/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA , Proteínas Ribossômicas/antagonistas & inibidores , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Ativação Transcricional , Regulação para Cima
3.
PLoS One ; 6(10): e25871, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22028794

RESUMO

Protein ubiquitination is a post-translational protein modification that regulates many biological conditions. Trip12 is a HECT-type E3 ubiquitin ligase that ubiquitinates ARF and APP-BP1. However, the significance of Trip12 in vivo is largely unknown. Here we show that the ubiquitin ligase activity of Trip12 is indispensable for mouse embryogenesis. A homozygous mutation in Trip12 (Trip12(mt/mt)) that disrupts the ubiquitin ligase activity resulted in embryonic lethality in the middle stage of development. Trip12(mt/mt) embryos exhibited growth arrest and increased expression of the negative cell cycle regulator p16. In contrast, Trip12(mt/mt) ES cells were viable. They had decreased proliferation, but maintained both the undifferentiated state and the ability to differentiate. Trip12(mt/mt) ES cells had increased levels of the BAF57 protein (a component of the SWI/SNF chromatin remodeling complex) and altered gene expression patterns. These data suggest that Trip12 is involved in global gene expression and plays an important role in mouse development.


Assuntos
Desenvolvimento Embrionário , Ubiquitina-Proteína Ligases/metabolismo , Animais , Ciclo Celular/genética , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/metabolismo , Feminino , Masculino , Camundongos , Mutação , Fenótipo , Estabilidade Proteica , Estrutura Terciária de Proteína , Transcriptoma , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética
4.
Cancer Sci ; 102(5): 1081-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21299717

RESUMO

Histone deacetylase inhibitors (HDACi) have been shown to exhibit anti-inflammatory activity, but their mechanism of action is poorly understood. Trichostatin A (TSA) and the cyclic tetrapeptide class inhibitor Ky-2 inhibit both lipopolysaccharide-induced tumor necrosis factor-α (TNF-α) production in rats and TNF-α-induced expression of inflammatory genes in HeLa cells. We assessed the molecular mechanism underlying TSA-induced anti-inflammatory activity by genetically dissecting activation of the nuclear factor-κB (NF-κB) pathway following stimulation with TNF-α. Trichostatin A did not inhibit degradation of IκBα, nuclear translocation and DNA binding of NF-κB; also, the drug did not affect transient expression from exogenous κB-reporter plasmids. However, endogenous expression of inflammatory cytokines such as interleukin-8 (IL-8) was greatly reduced, even in the absence of de novo protein synthesis, suggesting that HDACi directly inhibits NF-κB-induced transcription. Indeed, chromatin immunoprecipitation (ChIP) analysis showed that events related to transcriptional activation of the IL-8 gene region in response to TNF-α, including recruitment of RNA polymerase II (Pol II), were compromised in the presence of TSA. These data indicate that HDAC activity is required for the efficient initiation and/or elongation of inflammatory gene transcription mediated by NF-κB.


Assuntos
Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Inflamação/metabolismo , NF-kappa B/biossíntese , RNA Polimerase II/metabolismo , Animais , Western Blotting , Imunoprecipitação da Cromatina , Imunofluorescência , Células HeLa , Humanos , Ácidos Hidroxâmicos/farmacologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica
5.
Biochem Biophys Res Commun ; 404(1): 166-72, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21108932

RESUMO

Hepatic gluconeogenesis is crucial for glucose homeostasis. Although sirtuin 1 (Sirt1) is implicated in the regulation of gluconeogenesis in the liver, the effects of other histone deacetylases (HDAC) on gluconeogenesis are unclear. The aim of this study was to identify the role of class I HDACs in hepatic gluconeogenesis. In HepG2 cells and the liver of mice, the expressions of phosphoenol pyruvate carboxykinase (PEPCK) and hepatocyte nuclear factor 4α (HNF4α) were significantly decreased by treatment with a newly designed class I HDAC inhibitor, Ky-2. SiRNA knockdown of HDAC1 expression, but not of HDAC2 or HDAC3, in HepG2 cells decreased PEPCK and HNF4α expression. In HepG2 cells, insulin-stimulated phosphorylation of Akt and forkhead box O 1 (FoxO1) was increased by Ky-2. Pyruvate tolerance tests in Ky-2-treated high-fat-diet (HFD)-fed mice showed a marked reduction in blood glucose compared with vehicle-treated HFD mice. These data suggest that class I HDACs increase HNF4α protein expression and the transcriptional activity of FoxO1, followed by the induction of PEPCK mRNA expression and gluconeogenesis in liver.


Assuntos
Gluconeogênese , Glucose/metabolismo , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 1/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Fígado/metabolismo , Peptídeos Cíclicos/farmacologia , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Linhagem Celular Tumoral , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Fator 4 Nuclear de Hepatócito/antagonistas & inibidores , Histona Desacetilase 1/genética , Humanos , Insulina/farmacologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfoenolpiruvato Carboxiquinase (GTP)/antagonistas & inibidores , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Fosforilação , RNA Interferente Pequeno/genética , Transcrição Gênica
6.
Cancer Sci ; 101(11): 2483-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20726856

RESUMO

Spliceostatin A (SSA) is a methylated derivative of an antitumor natural product FR901464, which specifically binds and inhibits the SF3b spliceosome sub-complex. To investigate the selective antitumor activity of SSA, we focused on the regulation of vascular endothelial growth factor (VEGF) mRNA, since VEGF is a key regulatory component in tumor angiogenesis and known for the intricate regulation of mRNA processing, such as alternative splicing. We found that in HeLa cells SSA reduced the amount of both mRNA and protein of VEGF. Spliceostatin A not only inhibited the splicing reaction of VEGF pre-mRNA but also reduced the total amount of VEGF's transcripts, while SSA affected GAPDH mRNA to a lesser extent. Given a significant reduction in VEGF gene expression, SSA was expected to possess anti-angiogenic activity in vivo. Indeed, SSA inhibited cancer cell-derived angiogenesis in vivo in a chicken chorioallantoic membrane (CAM) assay. The inhibition of angiogenesis with SSA was abolished by addition of exogenous VEGF. We also performed global gene expression analyses of HeLa cells and found that the expression levels of 38% of total genes including VEGF decreased to <50% of the basal levels following 16 h of SSA treatment. These results suggest that the global interference of gene expression including VEGF in tumor cells is at least one of the mechanisms by which SSA (or FR901464) exhibits its strong antitumor activity.


Assuntos
Inibidores da Angiogênese/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neovascularização Patológica/genética , Piranos/farmacologia , Compostos de Espiro/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Inibidores da Angiogênese/química , Animais , Western Blotting , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Estrutura Molecular , Neovascularização Patológica/metabolismo , Neovascularização Patológica/prevenção & controle , Análise de Sequência com Séries de Oligonucleotídeos , Piranos/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Compostos de Espiro/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Mol Biol Cell ; 20(17): 3801-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19570909

RESUMO

Genomic instability in colorectal cancer is categorized into two distinct classes: chromosome instability (CIN) and microsatellite instability (MSI). MSI is the result of mutations in the mismatch repair (MMR) machinery, whereas CIN is often thought to be associated with a disruption in the APC gene. Clinical data has recently shown the presence of heterozygous mutations in ATR and Chk1 in human cancers that exhibit MSI, suggesting that those mutations may contribute to tumorigenesis. To determine whether reduced activity in the DNA damage checkpoint pathway would cooperate with MMR deficiency to induce CIN, we used siRNA strategies to partially decrease the expression of ATR or Chk1 in MMR-deficient colorectal cancer cells. The resultant cancer cells display a typical CIN phenotype, as characterized by an increase in the number of chromosomal abnormalities. Importantly, restoration of MMR proficiency completely inhibited induction of the CIN phenotype, indicating that the combination of partial checkpoint blockage and MMR deficiency is necessary to trigger CIN. Moreover, disruption of ATR and Chk1 in MMR-deficient cells enhanced the sensitivity to treatment with the commonly used colorectal chemotherapeutic compound, 5-fluorouracil. These results provide a basis for the development of a combination therapy for those cancer patients.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Instabilidade Cromossômica , Neoplasias Colorretais/genética , Reparo de Erro de Pareamento de DNA , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Centrossomo/metabolismo , Quinase 1 do Ponto de Checagem , Neoplasias Colorretais/tratamento farmacológico , Quebras de DNA de Cadeia Dupla , Fluoruracila/uso terapêutico , Humanos , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
8.
Mol Cell Biol ; 25(22): 9910-9, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16260606

RESUMO

In response to DNA damage or replication stress, the protein kinase ATR is activated and subsequently transduces genotoxic signals to cell cycle control and DNA repair machinery through phosphorylation of a number of downstream substrates. Very little is known about the molecular mechanism by which ATR is activated in response to genotoxic insults. In this report, we demonstrate that protein phosphatase 5 (PP5) is required for the ATR-mediated checkpoint activation. PP5 forms a complex with ATR in a genotoxic stress-inducible manner. Interference with the expression or the activity of PP5 leads to impairment of the ATR-mediated phosphorylation of hRad17 and Chk1 after UV or hydroxyurea treatment. Similar results are obtained in ATM-deficient cells, suggesting that the observed defect in checkpoint signaling is the consequence of impaired functional interaction between ATR and PP5. In cells exposed to UV irradiation, PP5 is required to elicit an appropriate S-phase checkpoint response. In addition, loss of PP5 leads to premature mitosis after hydroxyurea treatment. Interestingly, reduced PP5 activity exerts differential effects on the formation of intranuclear foci by ATR and replication protein A, implicating a functional role for PP5 in a specific stage of the checkpoint signaling pathway. Taken together, our results suggest that PP5 plays a critical role in the ATR-mediated checkpoint activation.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Nucleares/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Quinase 1 do Ponto de Checagem , DNA/química , Dano ao DNA , Reparo do DNA , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Células HeLa , Humanos , Hidroxiureia/farmacologia , Imunoprecipitação , Microscopia de Fluorescência , Mitose , Oligonucleotídeos/química , Fosforilação , Plasmídeos/metabolismo , Proteínas Quinases/metabolismo , RNA Interferente Pequeno/metabolismo , Proteína de Replicação A/metabolismo , Fase S , Transdução de Sinais , Raios Ultravioleta
9.
Cancer Res ; 65(19): 8853-60, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16204056

RESUMO

Cyclophilin A (CypA) was recently reported to be overexpressed in non-small-cell lung cancer, and represents a potentially novel therapeutic target. To determine the role of CypA in oncogenesis, stable RNA interference (RNAi)-mediated knockdown of CypA was established in two non-small-cell lung cancer cell lines (ADLC-5M2 and LC-103H), and these cells were grown as xenografts in severe combined immunodeficient mice. Tumor cell proliferation, apoptosis, and angiogenesis were measured by Ki67, terminal deoxyribonucleotidyl transferase-mediated dUTP nick-end labeling, and CD31 immunohistochemistry, respectively. Tumor glucose metabolism was assessed by fluorodeoxyglucose positron emission tomography imaging. Knockdown of CypA correlated in vivo with slower growth, less fluorodeoxyglucose uptake, decreased proliferation, and a greater degree of apoptosis in the tumors. These results establish the relevance of CypA to tumor growth in vivo, specifically to proliferation and apoptosis. Elucidation of the precise role of CypA in these pathways may lead to new targeted therapies for lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Ciclofilina A/antagonistas & inibidores , Neoplasias Pulmonares/patologia , Interferência de RNA , Animais , Apoptose/fisiologia , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Ciclofilina A/biossíntese , Ciclofilina A/genética , Fluordesoxiglucose F18/farmacocinética , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Neovascularização Patológica/metabolismo , Transfecção
10.
EMBO J ; 23(15): 3164-74, 2004 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-15282542

RESUMO

The ataxia-telangiectasia mutated and rad3-related (ATR) kinase orchestrates cellular responses to DNA damage and replication stress. Complete loss of ATR function leads to chromosomal instability and cell death. However, heterozygous ATR mutations are found in human cancers with microsatellite instability, suggesting that ATR haploinsufficiency contributes to tumorigenesis. To test this possibility, we generated human cell line and mouse model systems in which a single ATR allele was inactivated on a mismatch repair (MMR)-deficient background. Monoallelic ATR gene targeting in MLH1-deficient HCT 116 colon carcinoma cells resulted in hypersensitivity to genotoxic stress accompanied by dramatic increases in fragile site instability, and chromosomal amplifications and rearrangements. The ATR(+/-) HCT 116 cells also displayed compromised activation of Chk1, an important downstream target for ATR. In complementary studies, we demonstrated that mice bearing the same Atr(+/-)/Mlh1(-/-) genotype were highly prone to both embryonic lethality and early tumor development. These results demonstrate that MMR proteins and ATR functionally interact during the cellular response to genotoxic stress, and that ATR serves as a haploinsufficient tumor suppressor in MMR-deficient cells.


Assuntos
Pareamento Incorreto de Bases/genética , Proteínas de Ciclo Celular/metabolismo , Reparo do DNA/genética , Dosagem de Genes , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Transporte , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular , Instabilidade Cromossômica , Aberrações Cromossômicas , DNA/genética , DNA/metabolismo , Amplificação de Genes/genética , Genótipo , Humanos , Cariotipagem , Camundongos , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
11.
Cancer Res ; 62(17): 4916-21, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12208741

RESUMO

FK228 is a histone deacetylase (HDAC) inhibitor, the molecular mechanism of inhibition of which has been unknown. Here we show that reduction of an intramolecular disulfide bond of FK228 greatly enhanced its inhibitory activity and that the disulfide bond was rapidly reduced in cells by cellular reducing activity involving glutathione. Computer modeling suggests that one of the sulfhydryl groups of the reduced form of FK228 (redFK) interacts with the active-site zinc, preventing the access of the substrate. HDAC1 and HDAC2 were more strongly inhibited by redFK than HDAC4 and HDAC6. redFK was less active than FK228 in inhibiting in vivo HDAC activity, due to rapid inactivation in medium and serum. Thus, FK228 serves as a stable prodrug to inhibit class I enzymes and is activated by reduction after uptake into the cells. The glutathione-mediated activation also implicates its clinical usefulness for counteracting glutathione-mediated drug resistance in chemotherapy.


Assuntos
Antibacterianos/farmacologia , Antibióticos Antineoplásicos/farmacologia , Depsipeptídeos , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Peptídeos Cíclicos , Pró-Fármacos/farmacologia , Sequência de Aminoácidos , Antibacterianos/química , Antibacterianos/farmacocinética , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacocinética , Sítios de Ligação , Biotransformação , Estabilidade de Medicamentos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Glutationa/metabolismo , Células HeLa , Humanos , Isoenzimas/antagonistas & inibidores , Modelos Moleculares , Dados de Sequência Molecular , Oxirredução , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...