Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Biosci (Landmark Ed) ; 26(5): 76-84, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-34027651

RESUMEN

Recently, Up-frameshift protein 1 (UPF1) is reported to be downregulated in various cancers and its low expression is closely correlated with poor prognosis. UPF1 is well known as a master regulator of nonsense-mediated mRNA decay (NMD), which serves as a highly conserved mRNA surveillance process protecting cells from aberrant toxic transcripts. Due to dysfunction of UPF1, NMD fails to proceed, which contributes to tumor initiation and progression. This review shows a brief summary of the aberrant expression, functional roles and molecular mechanisms of UPF1 during tumorigenesis. Increasing evidence has indicated that UPF1 could serve as a potential biomarker for cancer diagnosis and treatment for future clinical applications in cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias/metabolismo , ARN Helicasas/biosíntesis , ARN Helicasas/genética , Transactivadores/biosíntesis , Transactivadores/genética , Empalme Alternativo , Animales , Biomarcadores de Tumor/genética , Carcinogénesis , Progresión de la Enfermedad , Regulación hacia Abajo , Epigénesis Genética , Genómica , Humanos , Ratones , Neoplasias/genética , Degradación de ARNm Mediada por Codón sin Sentido , Pronóstico , ARN Mensajero/metabolismo , Transducción de Señal
2.
Acta Pharmacol Sin ; 42(11): 1900-1912, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33536604

RESUMEN

Ras-GTPase activating SH3 domain-binding protein 1 (G3BP1) is a multifunctional binding protein involved in the development of a variety of human cancers. However, the role of G3BP1 in breast cancer progression remains largely unknown. In this study, we report that G3BP1 is upregulated and correlated with poor prognosis in breast cancer. Overexpression of G3BP1 promotes breast cancer cell proliferation by stimulating ß-catenin signaling, which upregulates a number of proliferation-related genes. We further show that G3BP1 improves the stability of ß-catenin by inhibiting its ubiquitin-proteasome degradation rather than affecting the transcription of ß-catenin. Mechanistically, elevated G3BP1 interacts with and inactivates GSK-3ß to suppress ß-catenin phosphorylation and degradation. Disturbing the G3BP1-GSK-3ß interaction accelerates the degradation of ß-catenin, impairing the proliferative capacity of breast cancer cells. Our study demonstrates that the regulatory mechanism of the G3BP1/GSK-3ß/ß-catenin axis may be a potential therapeutic target for breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proliferación Celular/fisiología , ADN Helicasas/biosíntesis , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/biosíntesis , ARN Helicasas/biosíntesis , Proteínas con Motivos de Reconocimiento de ARN/biosíntesis , beta Catenina/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , beta Catenina/antagonistas & inhibidores
3.
J Clin Invest ; 129(3): 1015-1029, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30688659

RESUMEN

MAPK4 is an atypical MAPK. Currently, little is known about its physiological function and involvement in diseases, including cancer. A comprehensive analysis of 8887 gene expression profiles in The Cancer Genome Atlas (TCGA) revealed that MAPK4 overexpression correlates with decreased overall survival, with particularly marked survival effects in patients with lung adenocarcinoma, bladder cancer, low-grade glioma, and thyroid carcinoma. Interestingly, human tumor MAPK4 overexpression also correlated with phosphorylation of AKT, 4E-BP1, and p70S6K, independent of the loss of PTEN or mutation of PIK3CA. This led us to examine whether MAPK4 activates the key metabolic, prosurvival, and proliferative kinase AKT and mTORC1 signaling, independent of the canonical PI3K pathway. We found that MAPK4 activated AKT via a novel, concerted mechanism independent of PI3K. Mechanistically, MAPK4 directly bound and activated AKT by phosphorylation of the activation loop at threonine 308. It also activated mTORC2 to phosphorylate AKT at serine 473 for full activation. MAPK4 overexpression induced oncogenic outcomes, including transforming prostate epithelial cells into anchorage-independent growth, and MAPK4 knockdown inhibited cancer cell proliferation, anchorage-independent growth, and xenograft growth. We concluded that MAPK4 can promote cancer by activating the AKT/mTOR signaling pathway and that targeting MAPK4 may provide a novel therapeutic approach for cancer.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Experimentales/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Helicasas/biosíntesis , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Activación Enzimática , Células HCT116 , Humanos , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Células PC-3 , Proteínas Proto-Oncogénicas c-akt/genética , ARN Helicasas/genética , Serina-Treonina Quinasas TOR/genética
4.
Hum Pathol ; 84: 213-220, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30339968

RESUMEN

DHX15 is an outstanding member of the DEAH-box RNA helicase family. A few studies suggest that DHX15 contributes to carcinogenesis in several tumor cell lines. However, whether DHX15 acts as an oncogene or tumor suppressor and its association with hepatocellular carcinoma (HCC) prognosis are still poorly understood. To address this question, we used immunohistochemistry to evaluate DHX15 expression patterns and their association with clinicopathological factors and the prognosis of patients with HCC. Our results showed that DHX15 expression was significantly higher in cancerous tissues than that in nontumor tissues (P < .0001). DHX15 expression in HCC patients was associated with differentiation status (P = .018), tumor number (P = .048), intrahepatic or extrahepatic metastasis (P = .001), serum α-fetoprotein (P = .006), hepatitis B virus level (P = .018), and recurrence (P < .001). In addition, the survival analysis revealed that the DHX15-high group had significantly decreased overall survival time (P = .004) and lower 1-year survival rates (P = .002) compared with the DHX15-low group. Furthermore, multivariate analysis identified DHX15 expression as an independent factor associated with poor prognosis in HCC (P = .036). In summary, these findings demonstrate, for the first time, that DHX15 is significantly upregulated in HCC and its high expression was correlated with poor prognosis, suggesting its pivotal role in the progression of HCC. The present results suggest that DHX15 may serve as a potential prognostic biomarker for HCC patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , ARN Helicasas/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/mortalidad , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/mortalidad , Masculino , Persona de Mediana Edad , Pronóstico , Regulación hacia Arriba , Adulto Joven
5.
Methods ; 126: 54-65, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28723582

RESUMEN

The Dicer family of ribonucleases plays a key role in small RNA-based regulatory pathways by generating short dsRNA fragments that modulate expression of endogenous genes, or protect the host from invasive nucleic acids. Beginning with its initial discovery, biochemical characterization of Dicer has provided insight about its catalytic properties. However, a comprehensive understanding of how Dicer's domains contribute to substrate-specific recognition and catalysis is lacking. One reason for this void is the lack of high-resolution structural information for a metazoan Dicer in the apo- or substrate-bound state. Both biochemical and structural studies are facilitated by large amounts of highly purified, active protein, and Dicer enzymes have historically been recalcitrant to overexpression and purification. Here we describe optimized procedures for the large-scale expression of Dicer in baculovirus-infected insect cells. We then outline a three-step protocol for the purification of large amounts (3-4mg of Dicer per liter of insect cell culture) of highly purified and active Dicer protein, suitable for biochemical and structural studies. Our methods are general and are extended to enable overexpression, purification and biochemical characterization of accessory dsRNA binding proteins that interact with Dicer and modulate its catalytic activity.


Asunto(s)
Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/aislamiento & purificación , ARN Helicasas/biosíntesis , ARN Helicasas/aislamiento & purificación , ARN Bicatenario/biosíntesis , ARN Bicatenario/aislamiento & purificación , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/aislamiento & purificación , Ribonucleasa III/biosíntesis , Ribonucleasa III/aislamiento & purificación , Animales , Baculoviridae , Fenómenos Bioquímicos/fisiología , Proteínas de Drosophila/genética , Drosophila melanogaster , Expresión Génica , ARN Helicasas/genética , ARN Bicatenario/genética , Proteínas de Unión al ARN/genética , Ribonucleasa III/genética , Células Sf9
6.
Biomed Pharmacother ; 92: 365-372, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28554132

RESUMEN

Although the roles of Up-frameshift 1 (UPF1) in hepatocellular carcinoma (HCC) have been partly revealed, the detailed mechanisms remain poorly understood. Here, quantitative real-time PCR (qRT-PCR) and immunohistochemistry assays indicated that UPF1 expression was decreased in HCC tissues compared to the corresponding adjacent tissues, and was negatively correlated with MRP2/ABCC2 expression. Cell viability and apoptosis analyses showed that overexpression of UPF1 enhanced HCC cell sensitivity to sorafenib treatment, while knockdown of UPF1 decreased the sensitivity. Additionally, ectopic expression of UPF1 suppressed the epithelial-mesenchymal transition (EMT) process and the generation of cells with stem cell properties. Mechanistically, UPF1 directly bound with ABCC2, increased nonsense-mediated mRNA decay (NMD) efficiency and thus led to downregualtion of ABCC2. Collectively, UPF1 functions as a tumor suppressor by preventing cancer stem cell (CSC)-like characteristics, inhibiting EMT process and enhancing chemotherapeutic sensitivity via inhibiting ABCC2 expression in HCC cells. These findings establish UPF1 as a potential therapeutic target for HCC patients.


Asunto(s)
Progresión de la Enfermedad , Neoplasias Hepáticas/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , ARN Helicasas/biosíntesis , ARN/biosíntesis , Transactivadores/biosíntesis , Células Hep G2 , Humanos , Neoplasias Hepáticas/prevención & control , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos
7.
Cell Death Dis ; 8(4): e2747, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28406479

RESUMEN

The role of the innate immune protein LGP2 (laboratory of genetics and physiology 2) in FMDV-infected cells remains unknown. Here, we demonstrate the antiviral role of LGP2 during FMDV infection. FMDV infection triggered LGP2 mRNA expression but reduced protein expression. Overexpression of LGP2 suppressed FMDV replication, and the inflammatory response was significantly inhibited by LGP2 in virus-infected cells. The N-terminal DExDc and the C-terminal regulatory domain regions of LGP2 were essential for LGP2-mediated antiviral activity against FMDV. Disruption of RNA recognition by LGP2 is suggested to abolish completely LGP2-mediated antiviral activity against FMDV. FMDV leader protein (Lpro), as well as the 3Cpro and 2B proteins were determined to possess the ability to induce reduction of LGP2 protein expression. 2B-induced reduction of LGP2 was independent of cleavage of eukaryotic translation initiation factor 4 gamma; and the proteasomes, lysosomes or caspase-dependent pathways were not involved in this process. The C-terminal amino acids of 101-154 were essential for 2B-induced reduction of LGP2 and upregulation of inflammatory response. Direct interaction was demonstrated between LGP2 and 2B. Our results describe the antiviral role of LGP2 against FMDV and a novel antagonistic mechanism of FMDV that is mediated by 2B protein.


Asunto(s)
Virus de la Fiebre Aftosa/fisiología , Fiebre Aftosa , Regulación Enzimológica de la Expresión Génica/inmunología , Inmunidad Innata/fisiología , ARN Helicasas , Replicación Viral/fisiología , Animales , Cricetinae , Fiebre Aftosa/enzimología , Fiebre Aftosa/inmunología , Células HEK293 , Humanos , ARN Helicasas/biosíntesis , ARN Helicasas/inmunología , ARN Viral/inmunología , ARN Viral/metabolismo , Porcinos , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
8.
Gene ; 594(2): 183-189, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27593564

RESUMEN

Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus that can cause acute encephalitis with a high fatality rate. RNA interference (RNAi) is a powerful tool to silence gene expression and a potential therapy for virus infection. In this study, the antiviral ability of eight shRNA expression plasmids targeting different sites of the NS3 and NS4A genes of JEV was determined in BHK21 cells and mice. The pGP-NS3-3 and pGP-NS4A-4 suppressed 93.9% and 82.0% of JEV mRNA in cells, respectively. The virus titer in cells was reduced approximately 950-fold by pretreating with pGP-NS3-4, and 640-fold by pretreating with pGP-NS4A-4. The results of western blot and immunofluorescence analysis showed JEV E protein and viral load in cells were remarkably inhibited by shRNA expression plasmids. The viral load in brains of mice pretreated with pGP-NS3-4 or pGP-NS4A-4 were reduced approximately 2400-fold and 800-fold, respectively, and the survival rate of mice challenged with JEV were 70% and 50%, respectively. However, the antiviral ability of shRNA expression plasmids was decreased over time. This study indicates that RNAi targeting of the NS3 and NS4A genes of JEV can sufficiently inhibit the replication of JEV in vitro and in vivo, and NS3 and NS4A genes might be potential targets of molecular therapy for JEV infection.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/fisiología , Regulación Viral de la Expresión Génica/genética , Marcación de Gen/métodos , Interferencia de ARN , ARN Interferente Pequeño , Proteínas no Estructurales Virales , Replicación Viral/genética , Animales , Línea Celular , Cricetinae , Encefalitis Japonesa/genética , Encefalitis Japonesa/metabolismo , Encefalitis Japonesa/terapia , Ratones , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/biosíntesis , ARN Helicasas/genética , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , Serina Endopeptidasas/biosíntesis , Serina Endopeptidasas/genética , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/biosíntesis , Proteínas no Estructurales Virales/genética
9.
Hum Mol Genet ; 25(14): 2905-2922, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27179789

RESUMEN

The X-linked DEAD-box RNA helicase DDX3 (DDX3X) is a multifunctional protein that has been implicated in gene regulation, cell cycle control, apoptosis, and tumorigenesis. However, the precise physiological function of Ddx3x during development remains unknown. Here, we show that loss of Ddx3x results in an early post-implantation lethality in male mice. The size of the epiblast marked by Oct3/4 is dramatically reduced in embryonic day 6.5 (E6.5) Ddx3x-/Y embryos. Preferential paternal X chromosome inactivation (XCI) in extraembryonic tissues of Ddx3x heterozygous (Ddx3x-/+) female mice with a maternally inherited null allele leads to placental abnormalities and embryonic lethality during development. In the embryonic tissues, Ddx3x exhibits developmental- and tissue-specific differences in escape from XCI. Targeted Ddx3x ablation in the epiblast leads to widespread apoptosis and abnormal growth, which causes embryonic lethality in the Sox2-cre/+;Ddx3xflox/Y mutant around E11.5. The observation of significant increases in γH2AX and p-p53Ser15 indicates DNA damage, which suggests that loss of Ddx3x leads to higher levels of genome damage. Significant upregulation of p21WAF1/Cip1 and p15Ink4b results in cell cycle arrest and apoptosis in Ddx3x-deficient cells. These results have uncovered that mouse Ddx3x is essential for both embryo and extraembryonic development.


Asunto(s)
Desarrollo Embrionario/genética , Placentación/genética , ARN Helicasas/genética , Activación Transcripcional/genética , Animales , Apoptosis/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , ARN Helicasas DEAD-box , Daño del ADN/genética , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Humanos , Ratones , Embarazo , ARN Helicasas/biosíntesis , Inactivación del Cromosoma X/genética
10.
Stem Cells Dev ; 24(3): 372-83, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25219788

RESUMEN

The gene trap method for embryonic stem cells is an efficient method for identifying new genes that are involved in development. Using this method, we identified a novel gene called helicase family gene related to gastrulation (helG). Helicase family proteins regulate many systems in the body that are related to cell survival. HelG encodes a protein of 137 kDa, which contains a DExH helicase motif that is now named DHX30. HelG is strongly expressed in neural cells (ie, in the headfold, neural plate, neural tube, and brain) and somites during embryogenesis. Growing homozygous mutant embryos have neither differentiated somites nor brains. In these mutants, development was retarded by embryonic day 7.5 (E7.5), and the mutants died at E9.5. After the purification of HelG, an untwisting experiment was performed to confirm the helicase activity of HelG for DNA in vitro. We report for the first time that a helicase family gene is required for differentiation during embryogenesis; this gene might interact with polynucleotides to regulate some genes that are important for early development and has a structure similar to that of a human DExH box helicase.


Asunto(s)
Desarrollo Embrionario/genética , Células Madre Embrionarias/enzimología , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica , ARN Helicasas/genética , Secuencia de Aminoácidos , Animales , ADN Complementario/genética , Inducción Enzimática , Genes Letales , Genes Reporteros , Genes Sintéticos , Vectores Genéticos/genética , Estratos Germinativos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , ARN Helicasas/biosíntesis , ARN Helicasas/química , ARN Helicasas/deficiencia , Alineación de Secuencia , Homología de Secuencia de Aminoácido
11.
BMC Cancer ; 14: 246, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24708616

RESUMEN

BACKGROUND: Advanced squamous cervical cancer, one of the most commonly diagnosed cancers in women, still remains a major problem in oncology due to treatment failure and distant metastasis. Antitumor therapy failure is due to both intrinsic and acquired resistance; intrinsic resistance is often decisive for treatment response. In this study, we investigated the specific pathways and molecules responsible for baseline therapy failure in locally advanced squamous cervical cancer. METHODS: Twenty-one patients with locally advanced squamous cell carcinoma were enrolled in this study. Primary biopsies harvested prior to therapy were analyzed for whole human gene expression (Agilent) based on the patient's 6 months clinical response. Ingenuity Pathway Analysis was used to investigate the altered molecular function and canonical pathways between the responding and non-responding patients. The microarray results were validated by qRT-PCR and immunohistochemistry. An additional set of 24 formalin-fixed paraffin-embedded cervical cancer samples was used for independent validation of the proteins of interest. RESULTS: A 2859-gene signature was identified to distinguish between responder and non-responder patients. 'DNA Replication, Recombination and Repair' represented one of the most important mechanisms activated in non-responsive cervical tumors, and the 'Role of BRCA1 in DNA Damage Response' was predicted to be the most significantly altered canonical pathway involved in intrinsic resistance (p = 1.86E-04, ratio = 0.262). Immunohistological staining confirmed increased expression of BRCA1, BRIP1, FANCD2 and RAD51 in non-responsive compared with responsive advanced squamous cervical cancer, both in the initial set of 21 cervical cancer samples and the second set of 24 samples. CONCLUSIONS: Our findings suggest that FA/BRCA pathway plays an important role in treatment failure in advanced cervical cancer. The assessment of FANCD2, RAD51, BRCA1 and BRIP1 nuclear proteins could provide important information about the patients at risk for treatment failure.


Asunto(s)
Proteína BRCA1/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/biosíntesis , Neoplasias de Células Escamosas/genética , ARN Helicasas/biosíntesis , Recombinasa Rad51/biosíntesis , Neoplasias del Cuello Uterino/genética , Adulto , Anciano , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Análisis por Micromatrices , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias de Células Escamosas/tratamiento farmacológico , Neoplasias de Células Escamosas/patología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/patología
12.
Oncogene ; 32(39): 4634-45, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23108394

RESUMEN

FOXM1 is implicated in genotoxic drug resistance but its role and mechanism of action remain unclear. Here, we establish that γH2AX foci, indicative of DNA double-strand breaks (DSBs), accumulate in a time-dependent manner in the drug-sensitive MCF-7 cells but not in the resistant counterparts in response to epirubicin. We find that FOXM1 expression is associated with epirubicin sensitivity and DSB repair. Ectopic expression of FOXM1 can increase cell viability and abrogate DSBs sustained by MCF-7 cells following epirubicin, owing to an enhancement in repair efficiency. Conversely, alkaline comet and γH2AX foci formation assays show that Foxm1-null cells are hypersensitive to DNA damage, epirubicin and γ-irradiation. Furthermore, we find that FOXM1 is required for DNA repair by homologous recombination (HR) but not non-homologous end joining (NHEJ), using HeLa cell lines harbouring an integrated direct repeat green fluorescent protein reporter for DSB repair. We also identify BRIP1 as a direct transcription target of FOXM1 by promoter analysis and chromatin-immunoprecipitation assay. In agreement, depletion of FOXM1 expression by small interfering RNA downregulates BRIP1 expression at the protein and mRNA levels in MCF-7 and the epirubicin-resistant MCF-7 Epi(R) cells. Remarkably, the requirement for FOXM1 for DSB repair can be circumvented by reintroduction of BRIP1, suggesting that BRIP1 is an important target of FOXM1 in DSB repair. Indeed, like FOXM1, BRIP1 is needed for HR. These data suggest that FOXM1 regulates BRIP1 expression to modulate epirubicin-induced DNA damage repair and drug resistance.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/fisiología , Resistencia a Antineoplásicos/fisiología , Epirrubicina/farmacología , Factores de Transcripción Forkhead/fisiología , Proteínas de Neoplasias/fisiología , ARN Helicasas/fisiología , Reparación del ADN por Recombinación/fisiología , Animales , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Femenino , Fibroblastos , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/antagonistas & inhibidores , Rayos gamma , Histonas/análisis , Humanos , Células MCF-7/efectos de los fármacos , Células MCF-7/metabolismo , Células MCF-7/efectos de la radiación , Ratones , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , ARN Helicasas/biosíntesis , ARN Helicasas/genética , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , ARN Interferente Pequeño/farmacología , Tolerancia a Radiación , Proteínas Recombinantes de Fusión/fisiología
13.
Diabetes Metab Res Rev ; 27(8): 965-70, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22069294

RESUMEN

OBJECTIVE: The contribution of innate immunity responsible for beta-cell destruction in fulminant type 1 diabetes (FT1D) and slowly progressive insulin-dependent diabetes mellitus (SPIDDM) is unclear. RESEARCH DESIGN AND METHODS: Islet-cell expression of Toll-like receptors (TLRs) including TLR3 and TLR4, the cytoplasmic retinoic acid-inducible protein I (RIG-I)-like helicases, RIG-I, melanoma differentiation-associated gene-5 and laboratory of genetics and physiology 2 in the affected islets were studied immuno-histochemically on three pancreases obtained 2-5 days after the onset of FT1D and a pancreas from a patient with SPIDDM. RESULTS: Laboratory of genetics and physiology 2 and RIG-I strongly expressed in beta cells in all three FT1D pancreases infected with enterovirus (VP1 antigen). Melanoma differentiation-associated gene-5 was hyper-expressed in all subsets of islet cells including beta cells and alpha cells. TLR3 and TLR4 were expressed in mononuclear cells that infiltrated to islets. IFN-alpha/beta was strongly expressed in islet cells. In contrast, pancreas of a patient with SPIDDM, enterovirus and expression of innate immune receptors including RIG-I, melanoma differentiation-associated gene-5, hyperexpression of laboratory of genetics and physiology 2 and mononuclear cells, which were positive for TLR3 and TLR4, and infiltration to the islets were not detected. CONCLUSIONS: These findings demonstrate that retinoic acid-inducible protein I (RIG-I)-like helicases and TLRs play a crucial role on beta-cell destruction in enterovirus-induced FT1D. The presence of distinct mechanism(s) of slowly progressive beta-cell failure in SPIDDM was suggested.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Inmunidad Innata , Islotes Pancreáticos/patología , Páncreas/patología , Adolescente , Adulto , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/biosíntesis , Diabetes Mellitus Tipo 1/virología , Cetoacidosis Diabética/mortalidad , Progresión de la Enfermedad , Infecciones por Enterovirus/complicaciones , Humanos , Helicasa Inducida por Interferón IFIH1 , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Masculino , ARN Helicasas/biosíntesis , Receptores Inmunológicos , Receptores Toll-Like/biosíntesis , Proteínas Estructurales Virales/biosíntesis
14.
RNA Biol ; 8(5): 904-12, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21788736

RESUMEN

During Drosophila development, translational control plays a crucial role in regulating gene expression, and is particularly important during pre-patterning of the maturing oocyte. A critical step in translation initiation is the binding of the eukaryotic translation initiation factor 4E (eIF4E) to the mRNA cap structure, which ultimately leads to recruitment of the ribosome. d4EHP is a translational repressor that prevents translation initiation by out-competing eIF4E on the cap structure for a subset of mRNAs. However, only two examples of mRNAs subject to d4EHP translation repression in Drosophila are known. Here we show that the belle (bel) mRNA is translationally repressed by the d4EHP protein in the Drosophila ovary. Consistent with this regulation, d4EHP overexpression in the ovary phenocopies the bel mutant. We also provide evidence that the Bel protein binds to eIF4E and may itself function as a translation repressor protein, with bruno as a potential target for Bel repression in the oocyte. Bruno is known to repress the mRNA of the key oocyte axis determinant oskar (osk) during oogenesis, and we find that an increase in the level of Bruno protein in bel mutant ovaries is associated with a reduction in Osk protein. Overall, our data suggest that a translational regulatory network exists in which consecutive translational repression events act to correctly pattern the Drosophila oocyte.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Oocitos/fisiología , ARN Helicasas/genética , Animales , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Oogénesis/genética , Unión Proteica , Biosíntesis de Proteínas/genética , Caperuzas de ARN/metabolismo , ARN Helicasas/biosíntesis , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
15.
J Virol ; 85(16): 8403-12, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21680521

RESUMEN

The retinoic acid-inducible gene I (RIG-I)-like receptors (RLR) comprise three homologues: RIG-I, melanoma differentiation-associated gene 5 (MDA5), and laboratory of genetics and physiology 2 (LGP2). They activate the host interferon (IFN) system upon recognition of viral RNA pathogen-associated molecular patterns (PAMPs) in the cytoplasm. Bioinformatic analysis of the sequenced vertebrate genomes suggests that the cytosolic surveillance system is conserved in lower vertebrates, and recent functional studies have confirmed that RIG-I is important to fish antiviral immunity. In this study, we have identified MDA5 and LGP2 homologues from rainbow trout Oncorhynchus mykiss and an additional LGP2 variant with an incomplete C-terminal domain of RIG-I. Trout MDA5 and LGP2 were constitutively produced in fibroblast and macrophage cell lines and upregulated by poly(I:C), recombinant IFN, or infection by RNA viruses (viral hemorrhagic septicemia virus and salmon alphavirus) with a single-stranded positive or negative genome. Overexpression of MDA5 and LGP2 but not of the LGP2 variant resulted in significant accumulation of Mx transcripts in cultured cells, which correlated with a marked enhancement of protection against viral infection. These results demonstrate that both MDA5 and LGP2 are important RLRs in host surveillance against infection of both negative and positive viruses and that the LGP2 variant with a deletion of 54 amino acids at the C terminus acts as a negative regulator for LGP2-elicited antiviral signaling by competing for the viral RNA PAMPs. Interestingly, MDA5 expression was not affected by overexpressed LGP2 in transfected cells and vice versa, suggesting that they likely act in parallel as positive regulators for IFN production.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Enfermedades de los Peces/inmunología , Proteínas de Peces/metabolismo , Oncorhynchus mykiss , ARN Helicasas/metabolismo , Infecciones por Rhabdoviridae/veterinaria , Alphavirus/inmunología , Animales , Línea Celular , ARN Helicasas DEAD-box/biosíntesis , ARN Helicasas DEAD-box/genética , Fibroblastos/inmunología , Enfermedades de los Peces/metabolismo , Enfermedades de los Peces/virología , Proteínas de Peces/biosíntesis , Proteínas de Peces/genética , Interferón Tipo I/inmunología , Macrófagos/inmunología , Novirhabdovirus/inmunología , Poli I-C/inmunología , Reacción en Cadena de la Polimerasa , ARN Helicasas/biosíntesis , ARN Helicasas/genética , ARN Viral/inmunología , ARN Viral/metabolismo , Proteínas Recombinantes , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/metabolismo , Infecciones por Rhabdoviridae/virología , Transducción de Señal/fisiología
16.
J Gen Virol ; 91(Pt 1): 133-44, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19793904

RESUMEN

Bovine viral diarrhea virus (BVDV; genus Pestivirus) can exist as two biotypes, cytopathogenic (CP) and non-cytopathogenic (NCP). The CP form differs from NCP by the continual expression of free non-structural protein 3 (NS3). CP BVDV infection of cultured cells induces apoptosis, whereas NCP BVDV infection has been reported to block the induction of beta interferon (IFN-beta). To investigate the viral mechanisms underlying these effects, NS3 or NS2-3 proteins of NCP and CP BVDV biotypes, together with the cognate NS3 co-factor NS4A, were expressed in cells, and their effect on apoptosis and induction of IFN-beta was investigated. Expression of NS3/4A resulted in increased activity of caspase-9 and caspase-3, indicating induction of the intrinsic apoptosis pathway. Mutational analysis revealed that a protease-inactive NS3/4A was unable to induce apoptosis, suggesting that NS3 protease activity is required for initiation of apoptosis during CP BVDV infection. The ability of NS2-3 to modulate activation of the IFN-beta promoter was also investigated. These studies confirmed that, unlike the related hepatitis C virus and GB virus-B, BVDV proteases are unable to inhibit TLR3- and RIG-I-dependent activation of the IFN-beta promoter. These data suggest that BVDV NS3/4A is responsible for regulating the levels of cellular apoptosis and provide new insights regarding the viral elements associated with CP biotype pathogenesis.


Asunto(s)
Apoptosis , Virus de la Diarrea Viral Bovina/patogenicidad , Interferón beta/genética , Péptido Hidrolasas/biosíntesis , Regiones Promotoras Genéticas , ARN Helicasas/biosíntesis , Proteínas no Estructurales Virales/biosíntesis , Sustitución de Aminoácidos/genética , Animales , Caspasa 3/biosíntesis , Caspasa 9/biosíntesis , Bovinos , Línea Celular , Efecto Citopatogénico Viral , Mutagénesis Sitio-Dirigida , Péptido Hidrolasas/genética , ARN Helicasas/genética , Proteínas no Estructurales Virales/genética
17.
Fish Shellfish Immunol ; 24(2): 223-33, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18166489

RESUMEN

RNA interference (RNAi) is an evolutionarily conserved mechanism by which double-stranded RNA (dsRNA) initiates post-transcriptional silencing of homologous genes. Here we report the amplification and characterisation of a full length cDNA from black tiger shrimp (Penaeus monodon) that encodes the bidentate RNAase III Dicer, a key component of the RNAi pathway. The full length of the shrimp Dicer (Pm Dcr1) cDNA is 7629bp in length, including a 5' untranslated region (UTR) of 130bp, a 3' UTR of 77bp, and an open reading frame of 7422bp encoding a polypeptide of 2473 amino acids with an estimated molecular mass of 277.895kDa and a predicted isoelectric point of 4.86. Analysis of the deduced amino acid sequence indicated that the mature peptide contains all the seven recognised functional domains and is most similar to the mosquito (Aedes aegypti) Dicer-1 sequence with a similarity of 34.6%. Quantitative RT-PCR analysis showed that Pm Dcr1 mRNA is most highly expressed in haemolymph and lymphoid organ tissues (P<0.05). However, there was no correlation between Pm Dcr1 mRNA levels in lymphoid organ and the viral genetic loads in shrimp naturally infected with gill-associated virus (GAV) and Mourilyan virus (P>0.05). Treatment with synthetic dsRNA corresponding to Pm Dcr1 sequence resulted in knock-down of Pm Dcr1 mRNA expression in both uninfected shrimp and shrimp infected experimentally with GAV. Knock-down of Pm Dcr1 expression resulted in more rapid mortalities and higher viral loads. These data demonstrated that Dicer is involved in antiviral defence in shrimp.


Asunto(s)
Expresión Génica/inmunología , Penaeidae/inmunología , Penaeidae/virología , ARN Helicasas/genética , Roniviridae/inmunología , Roniviridae/patogenicidad , Secuencia de Aminoácidos , Animales , ADN Complementario/química , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica/veterinaria , Orden Génico , Datos de Secuencia Molecular , Penaeidae/efectos de los fármacos , Filogenia , ARN Helicasas/análisis , ARN Helicasas/biosíntesis , ARN Bicatenario/farmacología , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Alineación de Secuencia/veterinaria , Homología de Secuencia de Aminoácido , Distribución Tisular , Carga Viral/veterinaria
18.
J Virol ; 80(22): 11255-64, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16971441

RESUMEN

Our previous studies using trans-complementation analysis of Kunjin virus (KUN) full-length cDNA clones harboring in-frame deletions in the NS3 gene demonstrated the inability of these defective complemented RNAs to be packaged into virus particles (W. J. Liu, P. L. Sedlak, N. Kondratieva, and A. A. Khromykh, J. Virol. 76:10766-10775). In this study we aimed to establish whether this requirement for NS3 in RNA packaging is determined by the secondary RNA structure of the NS3 gene or by the essential role of the translated NS3 gene product. Multiple silent mutations of three computer-predicted stable RNA structures in the NS3 coding region of KUN replicon RNA aimed at disrupting RNA secondary structure without affecting amino acid sequence did not affect RNA replication and packaging into virus-like particles in the packaging cell line, thus demonstrating that the predicted conserved RNA structures in the NS3 gene do not play a role in RNA replication and/or packaging. In contrast, double frameshift mutations in the NS3 coding region of full-length KUN RNA, producing scrambled NS3 protein but retaining secondary RNA structure, resulted in the loss of ability of these defective RNAs to be packaged into virus particles in complementation experiments in KUN replicon-expressing cells. Furthermore, the more robust complementation-packaging system based on established stable cell lines producing large amounts of complemented replicating NS3-deficient replicon RNAs and infection with KUN virus to provide structural proteins also failed to detect any secreted virus-like particles containing packaged NS3-deficient replicon RNAs. These results have now firmly established the requirement of KUN NS3 protein translated in cis for genome packaging into virus particles.


Asunto(s)
Biosíntesis de Proteínas , ARN Viral/metabolismo , Proteínas no Estructurales Virales/biosíntesis , Ensamble de Virus , Virus del Nilo Occidental/fisiología , Animales , Chlorocebus aethiops , Mutación del Sistema de Lectura , Genes Reporteros , Mutación , Conformación de Ácido Nucleico , ARN Helicasas/biosíntesis , ARN Helicasas/genética , ARN Viral/genética , Serina Endopeptidasas/biosíntesis , Serina Endopeptidasas/genética , Transcripción Genética , Células Vero , Proteínas no Estructurales Virales/genética , Ensamble de Virus/genética , Virus del Nilo Occidental/genética , beta-Galactosidasa/análisis , beta-Galactosidasa/genética
19.
Nucleic Acids Res ; 34(12): 3446-54, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16840531

RESUMEN

Expression of the cyanobacterial DEAD-box RNA helicase, crhR, is regulated in response to conditions, which elicit reduction of the photosynthetic electron transport chain. A combination of electrophoretic mobility shift assay (EMSA), DNA affinity chromatography and mass spectrometry identified that a LexA-related protein binds specifically to the crhR gene. Transcript analysis indicates that lexA and crhR are divergently expressed, with lexA and crhR transcripts accumulating differentially under conditions, which respectively oxidize and reduce the electron transport chain. In addition, expression of the Synechocystis lexA gene is not DNA damage inducible and its amino acid sequence lacks two of three residues required for activity of prototypical LexA proteins, which repress expression of DNA repair genes in a range of prokaryotes. A direct effect of recombinant LexA protein on crhR expression was confirmed from the observation that LexA reduces crhR expression in a linear manner in an in vitro transcription/translation assay. The results indicate that the Synechocystis LexA-related protein functions as a regulator of redox-responsive crhR gene expression, and not DNA damage repair genes.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , ARN Helicasas/genética , Proteínas Represoras/metabolismo , Serina Endopeptidasas/metabolismo , Synechocystis/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/fisiología , Sitios de Unión , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Oxidación-Reducción , Regiones Promotoras Genéticas , ARN Helicasas/biosíntesis , ARN Mensajero/biosíntesis , Rec A Recombinasas/biosíntesis , Rec A Recombinasas/genética , Proteínas Represoras/química , Serina Endopeptidasas/química , Serina Endopeptidasas/fisiología , Synechocystis/enzimología
20.
Cancer Res ; 66(11): 5574-81, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16740692

RESUMEN

RNA helicase A (RHA), a member of the DEXH box helicase family of proteins, is an integral component of protein complexes that regulate transcription and splicing. The EWS-FLI1 oncoprotein is expressed as a result of the chromosomal translocation t(11;22) that occurs in patients with the Ewing's sarcoma family of tumors (ESFT). Using phage display library screening, we identified an EWS-FLI1 binding peptide containing homology to RHA. ESFT cell lines and patient tumors highly expressed RHA. GST pull-down and ELISA assays showed that EWS-FLI1 specifically bound RHA fragment amino acids 630 to 1020, which contains the peptide region discovered by phage display. Endogenous RHA was identified in a protein complex with EWS-FLI1 in ESFT cell lines. Chromatin immunoprecipitation experiments showed both EWS-FLI1 and RHA bound to EWS-FLI1 target gene promoters. RHA stimulated the transcriptional activity of EWS-FLI1 regulated promoters, including Id2, in ESFT cells. In addition, RHA expression in mouse embryonic fibroblast cells stably transfected with EWS-FLI1 enhanced the anchorage-independent phenotype above that with EWS-FLI1 alone. These results suggest that RHA interacts with EWS-FLI1 as a transcriptional cofactor to enhance its function.


Asunto(s)
Autoantígenos/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , ARN Helicasas/metabolismo , Sarcoma de Ewing/metabolismo , Animales , Autoantígenos/biosíntesis , Autoantígenos/genética , Adhesión Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , ARN Helicasas DEAD-box , Humanos , Ratones , Ratones Desnudos , Proteínas de Neoplasias , Proteínas de Fusión Oncogénica/genética , Biblioteca de Péptidos , Péptidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteína Proto-Oncogénica c-fli-1/genética , ARN Helicasas/biosíntesis , ARN Helicasas/genética , Proteína EWS de Unión a ARN , Proteínas Recombinantes/metabolismo , Sarcoma de Ewing/enzimología , Sarcoma de Ewing/genética , Activación Transcripcional , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...