Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Arch Virol ; 155(6): 905-14, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20372944

RESUMO

The mechanisms responsible for virulence of influenza viruses in humans remain poorly understood. A prevailing hypothesis is that the highly pathogenic virus isolates cause a severe cytokinemia precipitating acute respiratory distress syndrome and multiple organ dysfunction syndrome. Cynomolgus macaques (Macaca fascicularis) infected with a human highly pathogenic avian influenza (HPAI) H5N1 virus isolate (A/Vietnam/1203/2004) or reassortants of human influenza virus A/Texas/36/91 (H1N1) containing genes from the 1918 pandemic influenza A (H1N1) virus developed severe pneumonia within 24 h postinfection. However, virus spread beyond the lungs was only detected in the H5N1 group, and signs of extrapulmonary tissue reactions, including microglia activation and sustained up-regulation of inflammatory markers, most notably hypoxia inducible factor-1alpha (HIF-1alpha), were largely limited to this group. Extrapulmonary pathology may thus contribute to the morbidities induced by H5N1 viruses.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Virus da Influenza A Subtipo H5N1/patogenicidade , Fígado/patologia , Microglia/imunologia , Infecções por Orthomyxoviridae/fisiopatologia , Animais , Citocinas/metabolismo , Humanos , Macaca fascicularis , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Sistema Respiratório/patologia , Regulação para Cima , Virulência
2.
J Med Primatol ; 38 Suppl 1: 17-23, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19863674

RESUMO

The National Primate Research Centers (NPRCs) established Working Groups (WGs) for developing resources and mechanisms to facilitate collaborations among non-human primate (NHP) researchers. Here we report the progress of the Genome Banking and the Genetics and Genomics WGs in developing resources to advance the exchange, analysis and comparison of NHP genetic and genomic data across the NPRCs. The Genome Banking WG has established a National NHP DNA bank comprising 1250 DNA samples from unrelated animals and family trios from the 10 NHP species housed within the NPRC system. The Genetics and Genomics WG is developing SNP arrays that will provide a uniform, highly informative, efficient and low-cost method for rhesus and long-tailed macaque genotyping across the eight NPRCs. This WG is also establishing a Biomedical Informatics Research Network-based portal for shared bioinformatics resources including vital statistics, genotype and population data and information on the National NHP DNA bank.


Assuntos
Genômica/organização & administração , Primatas/genética , Animais , National Institutes of Health (U.S.) , Estados Unidos
3.
J Virol ; 81(21): 11817-27, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17715226

RESUMO

We are still inadequately prepared for an influenza pandemic due to the lack of a vaccine effective for subtypes to which the majority of the human population has no prior immunity and which could be produced rapidly in sufficient quantities. There is therefore an urgent need to investigate novel vaccination approaches. Using a combination of genomic and traditional tools, this study compares the protective efficacy in macaques of an intrarespiratory live influenza virus vaccine produced by truncating NS1 in the human influenza A/Texas/36/91 (H1N1) virus with that of a conventional vaccine based on formalin-killed whole virus. After homologous challenge, animals in the live-vaccine group had greatly reduced viral replication and pathology in lungs and reduced upper respiratory inflammation. They also had lesser induction of innate immune pathways in lungs and of interferon-sensitive genes in bronchial epithelium. This postchallenge response contrasted with that shortly after vaccination, when more expression of interferon-sensitive genes was observed in bronchial cells from the live-vaccine group. This suggested induction of a strong innate immune response shortly after vaccination with the NS1-truncated virus, followed by greater maturity of the postchallenge immune response, as demonstrated with robust influenza virus-specific CD4+ T-cell proliferation, immunoglobulin G production, and transcriptional induction of T- and B-cell pathways in lung tissue. In conclusion, a single respiratory tract inoculation with an NS1-truncated influenza virus was effective in protecting nonhuman primates from homologous challenge. This protection was achieved in the absence of significant or long-lasting adverse effects and through induction of a robust adaptive immune response.


Assuntos
Sistema Imunitário/virologia , Orthomyxoviridae/genética , Orthomyxoviridae/metabolismo , Proteínas não Estruturais Virais/química , Animais , Biópsia , Sangue/virologia , Brônquios/patologia , Brônquios/virologia , Linfócitos T CD4-Positivos/metabolismo , Epitélio/virologia , Feminino , Regulação Viral da Expressão Gênica , Vírus da Influenza A Subtipo H1N1/metabolismo , Macaca , Masculino , Transcrição Gênica , Proteínas não Estruturais Virais/fisiologia
4.
J Virol ; 80(21): 10813-28, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16928763

RESUMO

Recent outbreaks of avian influenza in humans have stressed the need for an improved nonhuman primate model of influenza pathogenesis. In order to further develop a macaque model, we expanded our previous in vivo genomics experiments with influenza virus-infected macaques by focusing on the innate immune response at day 2 postinoculation and on gene expression in affected lung tissue with viral genetic material present. Finally, we sought to identify signature genes for early infection in whole blood. For these purposes, we infected six pigtailed macaques (Macaca nemestrina) with reconstructed influenza A/Texas/36/91 virus and three control animals with a sham inoculate. We sacrificed one control and two experimental animals at days 2, 4, and 7 postinfection. Lung tissue was harvested for pathology, gene expression profiling, and proteomics. Blood was collected for genomics every other day from each animal until the experimental endpoint. Gross and microscopic pathology, immunohistochemistry, viral gene expression by arrays, and/or quantitative real-time reverse transcription-PCR confirmed successful yet mild infections in all experimental animals. Genomic experiments were performed using macaque-specific oligonucleotide arrays, and high-throughput proteomics revealed the host response to infection at the mRNA and protein levels. Our data showed dramatic differences in gene expression within regions in influenza virus-induced lesions based on the presence or absence of viral mRNA. We also identified genes tightly coregulated in peripheral white blood cells and in lung tissue at day 2 postinoculation. This latter finding opens the possibility of using gene expression arrays on whole blood to detect infection after exposure but prior to onset of symptoms or shedding.


Assuntos
Influenza Humana/genética , Influenza Humana/virologia , Macaca nemestrina/genética , Macaca nemestrina/virologia , Animais , Modelos Animais de Doenças , Expressão Gênica , Perfilação da Expressão Gênica , Genes Virais , Genômica , Humanos , Imunidade Inata , Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Influenza Humana/imunologia , Influenza Humana/patologia , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Macaca nemestrina/imunologia , Masculino , Modelos Biológicos , Proteômica , Fatores de Tempo
6.
J Biol Chem ; 276(32): 30178-82, 2001 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-11487589

RESUMO

Double-stranded (ds) RNA, a common component of virus-infected cells, is a potent inducer of the type I interferon and other cellular genes. For identifying the full repertoire of human dsRNA-regulated genes, a cDNA microarray hybridization screening was conducted using mRNA from dsRNA-treated GRE cells. Because these cells lack all type I interferon genes, the possibility of gene induction by autocrine actions of interferon was eliminated. Our screen identified 175 dsRNA-stimulated genes (DSG) and 95 dsRNA-repressed genes. A subset of the DSGs was also induced by different inflammatory cytokines and viruses demonstrating interconnections among disparate signaling pathways. Functionally, the DSGs encode proteins involved in signaling, apoptosis, RNA synthesis, protein synthesis and processing, cell metabolism, transport, and structure. Induction of such a diverse family of genes by dsRNA has major implications in host-virus interactions and in the use of RNA(i) technology for functional ablation of specific genes.


Assuntos
Regulação da Expressão Gênica , Interferons/genética , RNA de Cadeia Dupla/metabolismo , RNA/metabolismo , Transdução de Sinais , Northern Blotting , Adesão Celular , Ciclo Celular , Linhagem Celular , Separação Celular , Citocinas/farmacologia , DNA Complementar/metabolismo , Regulação para Baixo , Citometria de Fluxo , Humanos , Cinética , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Tempo , Transcrição Gênica , Ativação Transcricional , Regulação para Cima
7.
Virology ; 284(1): 1-12, 2001 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-11352662

RESUMO

Interferons (IFNs) induce an antiviral state in the cell through complex and indirect mechanisms, which culminate in a direct inhibition of viral replication and stimulation of the host adaptive responses. Viruses often counteract with elaborate strategies to interfere with the induction as well as action of IFN effector molecules. This evolutionary battle between viruses and IFN components is a subject of intense research aimed at understanding the immunopathogenesis of viruses and the molecular basis of IFN signaling and action. In the case with hepatitis C virus (HCV), this may have profound implications for the therapeutic use of recombinant IFN in treating chronic hepatitis C. Depending on the subtype of HCV, current IFN-based treatment regimens are effective for only a small subset of chronic hepatitis C patients. Thus, one of the Holy Grails in HCV research is to understand the mechanisms by which the virus may evade IFN antiviral surveillance and establish persistent infection, which may eventually provide insights into new avenues for better antiviral therapy. Despite the lack of an efficient tissue culture system and an appropriate animal model for HCV infection, several mechanisms have been proposed based on clinical studies and in vitro experiments. This minireview focuses on the HCV NS5A nonstructural protein, which is implicated in playing a role in HCV tolerance to IFN treatment, possibly in part through its ability to inhibit the cellular IFN-induced PKR protein kinase.


Assuntos
Hepacivirus/imunologia , Interferons/imunologia , Proteínas não Estruturais Virais/fisiologia , Animais , Humanos , Modelos Biológicos , Transdução de Sinais , Ativação Transcricional , Proteínas não Estruturais Virais/genética
8.
J Virol ; 75(11): 5090-8, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11333890

RESUMO

The NS5A nonstructural protein of hepatitis C virus (HCV) has been shown to inhibit the cellular interferon (IFN)-induced protein kinase R (PKR). PKR mediates the host IFN-induced antiviral response at least in part by inhibiting mRNA translation initiation through phosphorylation of the alpha subunit of eukaryotic initiation factor 2 (eIF2alpha). We thus examined the effect of NS5A inhibition of PKR on mRNA translation within the context of virus infection by using a recombinant vaccinia virus (VV)-based assay. The VV E3L protein is a potent inhibitor of PKR. Accordingly, infection of IFN-pretreated HeLa S3 cells with an E3L-deficient VV (VVDeltaE3L) resulted in increased phosphorylation levels of both PKR and eIF2alpha. IFN-pretreated cells infected with VV in which the E3L locus was replaced with the NS5A gene (VVNS5A) displayed diminished phosphorylation of PKR and eIF2alpha in a transient manner. We also observed an increase in activation of p38 mitogen-activated protein kinase in IFN-pretreated cells infected with VVDeltaE3L, consistent with reports that p38 lies downstream of the PKR pathway. Furthermore, these cells exhibited increased phosphorylation of the cap-binding initiation factor 4E (eIF4E), which is downstream of the p38 pathway. Importantly, these effects were reduced in cells infected with VVNS5A. NS5A was also found to inhibit activation of the p38-eIF4E pathway in epidermal growth factor-treated cells stably expressing NS5A. NS5A-induced inhibition of eIF2alpha and eIF4E phosphorylation may exert counteracting effects on mRNA translation. Indeed, IFN-pretreated cells infected with VVNS5A exhibited a partial and transient restoration of cellular and viral mRNA translation compared with IFN-pretreated cells infected with VVDeltaE3L. Taken together, these results support the role of NS5A as a PKR inhibitor and suggest a potential mechanism by which HCV might maintain global mRNA translation rate during early virus infection while favoring cap-independent translation of HCV mRNA during late infection.


Assuntos
Hepacivirus/patogenicidade , Biossíntese de Proteínas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas não Estruturais Virais/biossíntese , Autorradiografia , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 4E em Eucariotos , Vetores Genéticos , Células HeLa , Hepacivirus/química , Humanos , Immunoblotting , Interferons/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , RNA Polimerase Dependente de RNA/fisiologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Transfecção , Vaccinia virus/genética , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/farmacologia , Proteínas Virais/genética , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
9.
J Virol ; 75(9): 4321-31, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11287581

RESUMO

Influenza virus, the causative agent of the common flu, is a worldwide health problem with significant economic consequences. Studies of influenza virus biology have revealed elaborate mechanisms by which the virus interacts with its host cell as it inhibits the synthesis of cellular proteins, evades the innate antiviral response, and facilitates production of viral RNAs and proteins. With the advent of DNA array technology it is now possible to obtain a large-scale view of how viruses alter the environment within the host cell. In this study, the cellular response to influenza virus infection was examined by monitoring the steady-state mRNA levels for over 4,600 cellular genes. Infections with active and inactivated influenza viruses identified changes in cellular gene expression that were dependent on or independent of viral replication, respectively. Viral replication resulted in the downregulation of many cellular mRNAs, and the effect was enhanced with time postinfection. Interestingly, several genes involved in protein synthesis, transcriptional regulation, and cytokine signaling were induced by influenza virus replication, suggesting that some may play essential or accessory roles in the viral life cycle or the host cell's stress response. The gene expression pattern induced by inactivated viruses revealed induction of the cellular metallothionein genes that may represent a protective response to virus-induced oxidative stress. Genome-scale analyses of virus infections will help us to understand the complexities of virus-host interactions and may lead to the discovery of novel drug targets or antiviral therapies.


Assuntos
Perfilação da Expressão Gênica , Orthomyxoviridae/fisiologia , Replicação Viral , Células HeLa , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Orthomyxoviridae/metabolismo , Raios Ultravioleta
10.
Virology ; 278(2): 501-13, 2000 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-11118372

RESUMO

Phosphorylation of the nonstructural NS5A protein is highly conserved among hepatitis C virus (HCV) genotypes. However, the precise site or sites of phosphorylation of NS5A have not been determined, and the functional significance of phosphorylation remains unknown. Here, we showed by two-dimensional phosphopeptide mapping that a protein kinase or kinases present in yeast, insect, and mammalian cells phosphorylated a highly purified HCV genotype 1b NS5A from insect cells on identical serine residues. We identified a major phosphopeptide (corresponding to amino acids 2193-2212 of the HCV 1b polyprotein) by using negative-ion electrospray ionization-microcapillary high performance liquid chromatography-mass spectrometry. The elution time of the phosphopeptide determined by negative-ion electrospray ionization-mass spectrometry corresponded with the elution time of the majority of (32)P-label that was incorporated into the phosphopeptide by an in vitro kinase reaction. Subsequent analysis of the peak fraction by automated positive-ion electrospray ionization-tandem mass spectrometry revealed that Ser(2194) was the major phosphorylated residue on the phosphopeptide GpSPPSLASSSASQLSAPSLK. Substitution for Ser(2194) with Ala resulted in the concomitant disappearance of major in vivo phosphorylated peptides. Ser(2194) and surrounding amino acids are highly conserved in all HCV genotypes, suggesting NS5A phosphorylation at Ser(2194) may be an important mechanism for modulating NS5A biological functions.


Assuntos
Hepacivirus/genética , Serina , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Clonagem Molecular , Sequência Consenso , Sequência Conservada , Hepacivirus/metabolismo , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fosfopeptídeos/química , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Saccharomyces cerevisiae , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Spodoptera , Transfecção
11.
Microbiol Mol Biol Rev ; 64(2): 239-80, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10839817

RESUMO

As obligate intracellular parasites, viruses rely exclusively on the translational machinery of the host cell for the synthesis of viral proteins. This relationship has imposed numerous challenges on both the infecting virus and the host cell. Importantly, viruses must compete with the endogenous transcripts of the host cell for the translation of viral mRNA. Eukaryotic viruses have thus evolved diverse mechanisms to ensure translational efficiency of viral mRNA above and beyond that of cellular mRNA. Mechanisms that facilitate the efficient and selective translation of viral mRNA may be inherent in the structure of the viral nucleic acid itself and can involve the recruitment and/or modification of specific host factors. These processes serve to redirect the translation apparatus to favor viral transcripts, and they often come at the expense of the host cell. Accordingly, eukaryotic cells have developed antiviral countermeasures to target the translational machinery and disrupt protein synthesis during the course of virus infection. Not to be outdone, many viruses have answered these countermeasures with their own mechanisms to disrupt cellular antiviral pathways, thereby ensuring the uncompromised translation of virion proteins. Here we review the varied and complex translational programs employed by eukaryotic viruses. We discuss how these translational strategies have been incorporated into the virus life cycle and examine how such programming contributes to the pathogenesis of the host cell.


Assuntos
Células Eucarióticas/virologia , Regulação Viral da Expressão Gênica , Biossíntese de Proteínas , Regiões 3' não Traduzidas/metabolismo , Animais , Fator de Iniciação 2 em Eucariotos/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação Eucariótico 4G , Herpesviridae/fisiologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/terapia , Infecções por Herpesviridae/virologia , Humanos , Orthomyxoviridae/genética , Fatores de Iniciação de Peptídeos/metabolismo , Ribossomos/genética
13.
Cell Mol Life Sci ; 57(2): 311-22, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10766025

RESUMO

Tetratricopeptide repeats (TPRs) are loosely conserved 34-amino acid sequence motifs that have been shown to function as scaffolding structures to mediate protein-protein interactions. TPRs have been identified in a number of proteins with diverse functions and cellular locations. Recent studies suggest that individual TPR motifs can confer specificity in promoting homotypic and/or heterotypic interactions, often in a mutually exclusive manner. These features are best exemplified by the P58IPK protein, an influenza virus-activated cellular inhibitor of the PKR protein kinase, whose different TPR motifs mediate interactions with distinct proteins. P58IPK, which possesses cochaperone and oncogenic properties, represents a unique class of TPR proteins containing a J-domain. Here we review recent progress on the structural and functional characterization of P58IPK, and discuss the possible mechanisms by which P58IPK modulates PKR and induces tumorigenesis in view of present knowledge of TPR proteins and molecular chaperones.


Assuntos
Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Proteínas Oncogênicas/metabolismo , Sequências Repetitivas de Aminoácidos , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Proteínas de Choque Térmico HSP40 , Humanos , Interferons/fisiologia , Modelos Biológicos , Dados de Sequência Molecular , Proteínas Oncogênicas/química , Estrutura Terciária de Proteína , eIF-2 Quinase/metabolismo
15.
Virology ; 266(1): 8-16, 2000 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-10612655

RESUMO

Human immunodeficiency virus type 1 (HIV-1) infection alters the expression of host cell genes at both the mRNA and protein levels. To obtain a more comprehensive view of the global effects of HIV infection of CD4-positive T-cells at the mRNA level, we performed cDNA microarray analysis on approximately 1500 cellular cDNAs at 2 and 3 days postinfection (p.i.) with HIV-1. Host cell gene expression changed little at 2 days p.i., but at 3 days p.i. 20 cellular genes were identified as differentially expressed. Genes involved in T-cell signaling, subcellular trafficking, and transcriptional regulation, as well as several uncharacterized genes, were among those whose mRNAs were differentially regulated. These results support the hypothesis that HIV-1 infection alters expression of a broad array of cellular genes and provides a framework for future functional studies on the differentially expressed mRNA products.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Regulação da Expressão Gênica , HIV-1/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Linfócitos T CD4-Positivos/patologia , Linhagem Celular , DNA Complementar , Perfilação da Expressão Gênica , Humanos , Processamento de Imagem Assistida por Computador , RNA Mensageiro/metabolismo , Transcrição Gênica
16.
J Interferon Cytokine Res ; 19(6): 543-54, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10433354

RESUMO

Recent research has thrown a spotlight on the interferon (IFN)-induced PKR protein kinase, implicating it as an important effector of apoptosis induced by several cellular stress conditions, including viral infection, cytokine treatment, and growth factor deprivation. In this review, we summarize the evidence for the role of PKR as a death accomplice and discuss how PKR might promote cell demise in light of current knowledge of the molecular mechanisms of apoptosis. Given its new found role and its established antiviral function, it is no wonder that PKR is a popular target for viral evasion of the host defense. PKR-dependent apoptosis may offer a novel cell-death pathway for specific manipulation in therapeutic strategies against apoptosis-related diseases.


Assuntos
Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Interferons/farmacologia , eIF-2 Quinase/biossíntese , Animais , Indução Enzimática , Genes Supressores de Tumor , Humanos , Vírus/patogenicidade
17.
J Virol ; 73(8): 6506-16, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10400746

RESUMO

Hepatitis C virus (HCV) is prevalent worldwide and has become a major cause of liver dysfunction and hepatocellular carcinoma. The high prevalence of HCV reflects the persistent nature of infection and the large frequency of cases that resist the current interferon (IFN)-based anti-HCV therapeutic regimens. HCV resistance to IFN has been attributed, in part, to the function of the viral nonstructural 5A (NS5A) protein. NS5A from IFN-resistant strains of HCV can repress the PKR protein kinase, a mediator of the IFN-induced antiviral and apoptotic responses of the host cell and a tumor suppressor. Here we examined the relationship between HCV persistence and resistance to IFN therapy. When expressed in mammalian cells, NS5A from IFN-resistant HCV conferred IFN resistance to vesicular stomatitis virus (VSV), which normally is sensitive to the antiviral actions of IFN. NS5A blocked viral double-stranded RNA (dsRNA)-induced PKR activation and phosphorylation of eIF-2alpha in IFN-treated cells, resulting in high levels of VSV mRNA translation. Mutations within the PKR-binding domain of NS5A restored PKR function and the IFN-induced block to viral mRNA translation. The effects due to NS5A inhibition of PKR were not limited to the rescue of viral mRNA translation but also included a block in PKR-dependent host signaling pathways. Cells expressing NS5A exhibited defective PKR signaling and were refractory to apoptosis induced by exogenous dsRNA. Resistance to apoptosis was attributed to an NS5A-mediated block in eIF-2alpha phosphorylation. Moreover, cells expressing NS5A exhibited a transformed phenotype and formed solid tumors in vivo. Disruption of apoptosis and tumorogenesis required the PKR-binding function of NS5A, demonstrating that these properties may be linked to the IFN-resistant phenotype of HCV.


Assuntos
Antivirais/farmacologia , Apoptose , Hepacivirus/fisiologia , Interferon-alfa/farmacologia , Proteínas não Estruturais Virais/metabolismo , eIF-2 Quinase/metabolismo , Sítios de Ligação , Resistência Microbiana a Medicamentos , Fator de Iniciação 2 em Eucariotos/metabolismo , Células HeLa , Hepacivirus/efeitos dos fármacos , Humanos , Mutagênese , Fosforilação , Poli I-C , Biossíntese de Proteínas , RNA de Cadeia Dupla , RNA Mensageiro , RNA Viral , Proteínas não Estruturais Virais/genética , eIF-2 Quinase/antagonistas & inibidores
18.
Mol Cell Biol ; 19(7): 4757-65, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10373525

RESUMO

P58(IPK) is a tetratricopeptide repeat-containing cochaperone that is involved in stress-activated cellular pathways and that inhibits the activity of protein kinase PKR, a primary mediator of the antiviral and antiproliferative properties of interferon. To gain better insight into the molecular actions of P58(IPK), we generated NIH 3T3 cell lines expressing either wild-type P58(IPK) or a P58(IPK) deletion mutant, DeltaTPR6, that does not bind to or inhibit PKR. When treated with double-stranded RNA (dsRNA), DeltaTPR6-expressing cells exhibited a significant increase in eukaryotic initiation factor 2alpha phosphorylation and NF-kappaB activation, indicating a functional PKR. In contrast, both of these PKR-dependent events were blocked by the overexpression of wild-type P58(IPK). In addition, the P58(IPK) cell line, but not the DeltaTPR6 cell line, was resistant to dsRNA-induced apoptosis. Together, these findings demonstrate that P58(IPK) regulates dsRNA signaling pathways by inhibiting multiple PKR-dependent functions. In contrast, both the P58(IPK) and DeltaTPR6 cell lines were resistant to tumor necrosis factor alpha-induced apoptosis, suggesting that P58(IPK) may function as a more general suppressor of programmed cell death independently of its PKR-inhibitory properties. In accordance with this hypothesis, although PKR remained active in DeltaTPR6-expressing cells, the DeltaTPR6 cell line displayed a transformed phenotype and was tumorigenic in nude mice. Thus, the antiapoptotic function of P58(IPK) may be an important factor in its ability to malignantly transform cells.


Assuntos
Apoptose , Chaperonas Moleculares/metabolismo , Inibidores de Proteínas Quinases , RNA de Cadeia Dupla/metabolismo , Proteínas Repressoras/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , eIF-2 Quinase/metabolismo , Células 3T3 , Animais , Fator de Iniciação 2 em Eucariotos/metabolismo , Proteínas de Choque Térmico HSP40 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Chaperonas Moleculares/genética , Mutagênese , NF-kappa B/metabolismo , Fenótipo , Fosforilação , Poli I-C/metabolismo , Poli I-C/farmacologia , RNA de Cadeia Dupla/antagonistas & inibidores , Coelhos , Proteínas Repressoras/genética , Transformação Genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia
19.
Biotechniques ; 26(6): 1102-6, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10376149

RESUMO

The yeast three-hybrid system presents a valuable tool for detecting and analyzing RNA-protein interactions in vivo. A major drawback of the use of such a transcriptional reporter-based assay in a library screen is the frequent occurrence of false-positive results due to bait RNA-independent activation of the reporter gene. To minimize the isolation of false positives in three-hybrid library screens, we incorporated a rapid and simple procedure based on differential sensitivity to 5-fluoro-orotic acid. The technique effectively eliminates bait RNA-independent false positives and thus greatly enhances the efficiency of the yeast three-hybrid system.


Assuntos
Clonagem Molecular/métodos , DNA Fúngico/análise , Ácido Orótico/análogos & derivados , Leveduras/genética , DNA Complementar , Reações Falso-Positivas , Biblioteca Gênica , Testes Genéticos , RNA Fúngico/análise , RNA Mensageiro/análise , Sensibilidade e Especificidade
20.
Proc Natl Acad Sci U S A ; 96(12): 6694-9, 1999 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-10359774

RESUMO

To better understand regulation of eukaryotic protein synthesis, we studied cellular and viral mRNA translation in influenza virus-infected cells. Influenza virus infection results in a dramatic shut-off of cellular protein synthesis that is concomitant with selective viral mRNA translation. Earlier work showed that these events are mediated by viral and/or cellular factors binding to the 5' untranslated region (5' UTR) of viral mRNAs. To identify trans-acting cellular proteins responsible for selective viral protein synthesis, we employed the yeast three-hybrid system. Using the 5' UTR of the influenza virus nucleocapsid protein (NP) mRNA as bait, we identified the cellular RNA-recognition motif containing RNA-binding protein G-rich sequence factor 1 (GRSF-1) as a positive-acting translational regulatory factor. The in vivo yeast assay revealed GRSF-1 specifically bound to the NP 5' UTR but not select NP 5' UTR mutants or cellular RNA 5' UTRs. These data were confirmed by gel shift assays using recombinant GRSF-1. Importantly, recombinant GRSF-1 specifically stimulated translation of a NP 5' UTR-driven template in cell-free translation systems. Furthermore, translation efficiency of NP 5' UTR-driven templates was reduced markedly in GRSF-1-depleted HeLa cell extracts, but restored in GRSF-1-reconstituted extracts. GRSF-1 also stimulated translation of an NP 5' UTR-driven template in HeLa cell extracts that were depleted of essential factors by addition of RNA oligonucleotides representing the viral 5' UTR RNA. Taken together, these data document the functional demonstration of a cellular protein binding to influenza virus RNAs and, importantly, suggest that influenza virus may recruit GRSF-1 to the 5' UTR to ensure preferential translation of viral mRNAs in infected cells.


Assuntos
Orthomyxoviridae/genética , Proteínas de Ligação a Poli(A) , Biossíntese de Proteínas , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Regiões 5' não Traduzidas/genética , Sequência de Bases , Sistema Livre de Células , Células HeLa , Humanos , Influenza Humana/genética , Influenza Humana/metabolismo , Dados de Sequência Molecular , RNA Viral/genética , Proteínas de Ligação a RNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA