Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
1.
Arch Virol ; 155(6): 905-14, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20372944

RESUMEN

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.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Hígado/patología , Microglía/inmunología , Infecciones por Orthomyxoviridae/fisiopatología , Animales , Citocinas/metabolismo , Humanos , Macaca fascicularis , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Sistema Respiratorio/patología , Regulación hacia Arriba , Virulencia
2.
Science ; 257(5077): 1685-9, 1992 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-1382315

RESUMEN

The double-stranded RNA-dependent protein kinase (dsRNA-PK) is thought to be a key mediator of the antiviral and antiproliferative effects of interferons (IFNs). Studies examining the physiological function of the kinase suggest that it participates in cell growth and differentiation by regulating protein synthesis. Autophosphorylation and consequent activation of dsRNA-PK in vitro and in vivo result in phosphorylation of the alpha subunit of eukaryotic initiation factor-2 (eIF-2) and inhibition of protein synthesis. Expression of a functionally defective mutant of human dsRNA-PK in NIH 3T3 cells resulted in malignant transformation, suggesting that dsRNA-PK may function as a suppressor of cell proliferation and tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica , Interferones/farmacología , Mutación , Proteínas Quinasas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , División Celular , Línea Celular , Clonación Molecular , ADN/genética , Inducción Enzimática , Expresión Génica , Humanos , Immunoblotting , Ratones , Datos de Secuencia Molecular , Fosforilación , Proteínas Quinasas/química , Proteínas Quinasas/fisiología , Transfección , eIF-2 Quinasa
3.
Science ; 247(4947): 1216-9, 1990 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-2180064

RESUMEN

The tat-responsive region (TAR) of the human immunodeficiency virus-1 (HIV-1) exhibits a trans-inhibitory effect on translation in vitro by activating the interferon-induced 68-kilodalton protein kinase (p68 kinase). Productive infection by HIV-1 was shown to result in a significant decrease in the amount of cellular p68 kinase. The steady-state amount of p68 kinase was also reduced in interferon-treated HeLa cell lines stably expressing tat, as compared to the amount of the kinase in interferon-treated control HeLa cells. Thus, the potential translational inhibitory effects of the TAR RNA region mediated by activation of p68 kinase may be downregulated by tat during productive HIV-1 infection.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Productos del Gen tat/fisiología , Genes Virales , Genes tat , VIH-1/genética , Interferón Tipo I/farmacología , Proteínas Quinasas/genética , Transactivadores/fisiología , 2',5'-Oligoadenilato Sintetasa/genética , Regulación hacia Abajo , Inducción Enzimática , Células HeLa , Humanos , Técnicas de Inmunoadsorción , Peso Molecular , Proteínas Quinasas/biosíntesis , Transfección , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
4.
Science ; 257(5066): 103-6, 1992 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-1621083

RESUMEN

After observations that Macaca nemestrina were exceptionally susceptible to simian immunodeficiency virus and human immunodeficiency virus type-2 (HIV-2), studies of HIV-1 replication were initiated. Several strains of HIV-1, including a recent patient isolate, replicated in vitro in peripheral blood mononuclear cells (PBMCs) and in CD4-positive M. nemestrina lymphocytes in a CD4-dependent fashion. Eight animals were subsequently inoculated with either cell-associated or cell-free suspensions of HIV-1. All animals had HIV-1 isolated by cocultivation, had HIV-1 DNA in their PBMCs as shown by polymerase chain reaction, and experienced sustained seroconversion to a broad spectrum of HIV-1 proteins. Macaca nemestrina is an animal model of HIV-1 infections that provides opportunities for evaluating the pathogenesis of acute HIV-1 replication and candidate vaccines and therapies.


Asunto(s)
Genes gag , Infecciones por VIH/fisiopatología , VIH-1/fisiología , Macaca nemestrina/microbiología , Replicación Viral , Animales , Secuencia de Bases , Antígenos CD4/fisiología , Cisteína/metabolismo , Bases de Datos Factuales , Seropositividad para VIH , VIH-1/aislamiento & purificación , VIH-1/patogenicidad , Humanos , Linfocitos/inmunología , Linfocitos/fisiología , Metionina/metabolismo , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos , Sondas de Oligonucleótidos , Proteínas Virales/biosíntesis , Proteínas Virales/aislamiento & purificación
5.
J Med Primatol ; 38 Suppl 1: 17-23, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19863674

RESUMEN

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.


Asunto(s)
Genómica/organización & administración , Primates/genética , Animales , National Institutes of Health (U.S.) , Estados Unidos
6.
Microbiol Mol Biol Rev ; 64(2): 239-80, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10839817

RESUMEN

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.


Asunto(s)
Células Eucariotas/virología , Regulación Viral de la Expresión Génica , Biosíntesis de Proteínas , Regiones no Traducidas 3'/metabolismo , Animales , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Factor 4G Eucariótico de Iniciación , Herpesviridae/fisiología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/terapia , Infecciones por Herpesviridae/virología , Humanos , Orthomyxoviridae/genética , Factores de Iniciación de Péptidos/metabolismo , Ribosomas/genética
7.
Mol Cell Biol ; 1(9): 807-13, 1981 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9279393

RESUMEN

An adenovirus type 5 host range mutant (hr-1) located in region E1A and phenotypically defective in expressing viral messenger ribonucleic acid (RNA) from other early regions (Berk et al., Cell 17:935-944, 1979) was analyzed for accumulation of viral RNA in the presence of protein synthesis inhibitors. Nuclear RNA was transcribed from all early regions at the same rate, regardless of whether the drug was present or absent. As expected, low or undetectable levels of RNA were found in the cytoplasm of hr-1-infected cells compared with the wild-type adenovirus type 5 in the absence of drug. When anisomycin was added 30 min before hr-1 infection, cytoplasmic RNA was abundant from early regions E3 and E4 when assayed by filter hybridization. In accordance, early regions E3 and E4 viral messenger RNA species were detected by the S1 endonuclease mapping technique only in hr-1-infected cells that were treated with the drug. Similar results were obtained by in vitro translation studies. Together, these results suggest that this adenovirus type 5 mutant lacks a viral gene product necessary for accumulation of viral messenger RNA, but not for transcription. It is proposed that a cellular gene product serves as a negative regulator of viral messenger RNA accumulation at the posttranscriptional level.


Asunto(s)
Adenoviridae/genética , Regulación Viral de la Expresión Génica , Procesamiento Postranscripcional del ARN , ARN Viral/metabolismo , Transcripción Genética , Citoplasma/metabolismo , Células HeLa , Humanos , Mutagénesis , Biosíntesis de Proteínas , ARN Mensajero/metabolismo
8.
Mol Cell Biol ; 4(10): 2198-206, 1984 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-6095046

RESUMEN

Influenza virus infection has adverse effects on the metabolism of two representative RNA polymerase II transcripts in chicken embryo fibroblasts, those coding for beta-actin and for avian leukosis virus (ALV) proteins. Proviral ALV DNA was integrated into host cell DNA by prior infection with ALV. Within 1 h after influenza virus infection, the rate of transcription of beta-actin and ALV sequences decreased 40 to 60%, as determined by labeling the cells for 5 min with [3H]uridine and by in vitro, runoff assays with isolated nuclei. The transcripts that continued to be synthesized did not appear in the cytoplasm as mature mRNAs, and the kinetics of labeling of these transcripts strongly suggest that they were degraded in the nucleus. By S1 endonuclease assay, it was confirmed that nuclear ALV transcripts disappeared very early after infection, already decreasing ca. 80% by 1 h postinfection. A plausible explanation for this nuclear degradation is that the viral cap-dependent endonuclease in the nucleus cleaves the 5' ends of new polymerase II transcripts, rendering the resulting decapped RNAs susceptible to hydrolysis by cellular nucleases. In contrast to the nuclear transcripts, cytoplasmic beta-actin and ALV mRNAs, which are synthesized before infection, were more stable and did not decrease in amount until after 3 h postinfection. Similar stability of cytoplasmic host cell mRNAs was observed in infected HeLa cells, in which the levels of actin mRNA and two HeLa cell mRNAs (pHe 7 and pHe 28) remained at undiminished levels for 3 h of infection and decreased only slightly by 4.5 h postinfection. The cytoplasmic actin and pHe 7 mRNAs isolated from infected HeLa cells were shown to be translated in reticulocyte extracts in vitro, indicating that host mRNAs were not inactivated by a virus-induced modification. Despite the continued presence of high levels of functional host cell mRNAs, host cell protein synthesis was effectively shut off by about 3 h postinfection in both chicken embryo fibroblasts and HeLa cells. These results are consistent with the establishment of an influenza virus-specific translational system that selectively translates viral and not host mRNAs.


Asunto(s)
Infecciones por Orthomyxoviridae/enzimología , ARN Polimerasa II/metabolismo , Transcripción Genética , Actinas/genética , Animales , Virus de la Leucosis Aviar/metabolismo , Embrión de Pollo , Fibroblastos/metabolismo , Células HeLa/metabolismo , Humanos , Virus de la Influenza A , ARN Mensajero/metabolismo , Factores de Tiempo , Proteínas Virales/metabolismo
9.
Mol Cell Biol ; 18(5): 2431-43, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9566864

RESUMEN

The interferon (IFN)-induced, double-stranded RNA-activated protein kinase (PKR) mediates the antiviral and antiproliferative actions of IFN, in part, via its translational inhibitory properties. Previous studies have demonstrated that PKR forms dimers and that dimerization is likely to be required for activation and/or function. In the present study we used multiple approaches to examine the modulation of PKR dimerization. Deletion analysis with the lambda repressor fusion system identified a previously unrecognized site involved in PKR dimerization. This site comprised amino acids (aa) 244 to 296, which span part of the third basic region of PKR and the catalytic subdomains I and II. Using the yeast two-hybrid system and far-Western analysis, we verified the importance of this region for dimerization. Furthermore, coexpression of the 52-aa region alone inhibited the formation of full-length PKR dimers in the lambda repressor fusion and two-hybrid systems. Importantly, coexpression of aa 244 to 296 exerted a dominant-negative effect on wild-type kinase activity in a functional assay. Due to its role as a mediator of IFN-induced antiviral resistance, PKR is a target of viral and cellular inhibitors. Curiously, PKR aa 244 to 296 contain the binding site for a select group of specific inhibitors, including the cellular protein P58IPK. We demonstrated, utilizing both the yeast and lambda systems, that P58IPK, a member of the tetratricopeptide repeat protein family, can block kinase activity by preventing PKR dimerization. In contrast, a nonfunctional form of P58IPK lacking a TPR motif did not inhibit kinase activity or perturb PKR dimers. These results highlight a potential mechanism of PKR inhibition and define a novel class of PKR inhibitors. Finally, the data document the first known example of inhibition of protein kinase dimerization by a cellular protein inhibitor. On the basis of these results we propose a model for the regulation of PKR dimerization.


Asunto(s)
Proteínas de Unión al ADN , Proteínas Represoras/farmacología , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/metabolismo , Sitios de Unión , Análisis Mutacional de ADN , Dimerización , Interferones/farmacología , Modelos Biológicos , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Unión Proteica/efectos de los fármacos , ARN Bicatenario/metabolismo , Proteínas Recombinantes de Fusión , Proteínas Represoras/genética , Eliminación de Secuencia , Proteínas Virales , Proteínas Reguladoras y Accesorias Virales , eIF-2 Quinasa/genética
10.
Mol Cell Biol ; 14(4): 2331-42, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7511204

RESUMEN

PKR is a serine/threonine protein kinase induced by interferon treatment and activated by double-stranded RNAs. As a result of activation, PKR becomes autophosphorylated and catalyzes phosphorylation of the alpha subunit of protein synthesis eukaryotic initiation factor 2 (eIF-2). While studying the regulation of PKR in virus-infected cells, we found that a cellular 58-kDa protein (P58) was recruited by influenza virus to downregulate PKR and thus avoid the kinase's deleterious effects on viral protein synthesis and replication. We now report on the cloning, sequencing, expression, and structural analysis of the P58 PKR inhibitor, a 504-amino-acid hydrophilic protein. P58, expressed as a histidine fusion protein in Escherichia coli, blocked both the autophosphorylation of PKR and phosphorylation of the alpha subunit of eIF-2. Western blot (immunoblot) analysis showed that P58 is present not only in bovine cells but also in human, monkey, and mouse cells, suggesting the protein is highly conserved. Computer analysis revealed that P58 contains regions of homology to the DnaJ family of proteins and a much lesser degree of similarity to the PKR natural substrate, eIF-2 alpha. Finally, P58 contains nine tandemly arranged 34-amino-acid repeats, demonstrating that the PKR inhibitor is a member of the tetratricopeptide repeat family of proteins, the only member identified thus far with a known biochemical function.


Asunto(s)
Interferones/farmacología , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Represoras/metabolismo , eIF-2 Quinasa/antagonistas & inhibidores , Secuencia de Aminoácidos , Secuencia de Bases , Secuencia Conservada , Cartilla de ADN , Inducción Enzimática , Factor 2 Eucariótico de Iniciación/química , Factor 2 Eucariótico de Iniciación/metabolismo , Proteínas del Choque Térmico HSP40 , Datos de Secuencia Molecular , Peso Molecular , Mutagénesis Sitio-Dirigida , Oligodesoxirribonucleótidos , Sondas de Oligonucleótidos , Fosforilación , Mutación Puntual , Reacción en Cadena de la Polimerasa , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos , Proteínas Represoras/genética , Proteínas Represoras/aislamiento & purificación , Eliminación de Secuencia , Homología de Secuencia de Aminoácido , Transcripción Genética
11.
Mol Cell Biol ; 15(6): 3138-46, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7539103

RESUMEN

Recently we reported that introduction of catalytically inactive PKR molecules into NIH 3T3 cells causes malignant transformation and the development of tumors in nude mice. We have proposed that PKR may be a tumor suppressor gene possibly because of its translational inhibitory properties. We have now designed and characterized a number of PKR mutants encoding proteins that retain their catalytic competence but are mutated in their regulatory double-stranded RNA (dsRNA) binding domains (RBDs). RNA binding analysis revealed that PKR proteins either lacking or with point mutations in the first RBD (RBD-1) bound negligible amounts of dsRNA activator or adenovirus VAI RNA inhibitor. Despite the lack of binding, such variants remained functionally competent but were much less active than wild-type PKR. PKR variants completely lacking RBD-1 were largely unresponsive to dsRNA in activation assays but could be activated by heparin. To complement these studies, we evaluated the effects of point mutations in RBD-1 or the removal of either RBD-1 or RBD-2 on the proliferation rate of mouse 3T3 cells. We were unsuccessful at isolating stably transformed cells expressing RBD-1 point mutants or RBD-2-minus mutants. In contrast, NIH 3T3 cells, which constitutively expressed PKR proteins that lacked RBD-1, were selected. These cells displayed a transformed phenotype and caused tumors after inoculation in nude mice. Further, levels of endogenous eIF-2 alpha phosphorylation in RBD-1-minus cell lines were reduced, suggesting that such mutants act in a dominant negative manner to inhibit the function of endogenous PKR. These results emphasize the importance of RBD-1 in PKR control of cell growth and provide additional evidence for the critical role played by PKR in the regulation of malignant transformation.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Células 3T3 , Animales , Secuencia de Bases , Sitios de Unión/genética , Transformación Celular Neoplásica/genética , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Serina-Treonina Quinasas/metabolismo , ARN/antagonistas & inhibidores , ARN/metabolismo , eIF-2 Quinasa
12.
Mol Cell Biol ; 6(5): 1741-50, 1986 May.
Artículo en Inglés | MEDLINE | ID: mdl-3785177

RESUMEN

Selective translation of influenza viral mRNAs occurs after influenza virus superinfection of cells infected with the VAI RNA-negative adenovirus mutant dl331 (M. G. Katze, Y.-T. Chen, and R. M. Krug, Cell 37:483-490, 1984). Cell extracts from these doubly infected cells catalyze the initiation of essentially only influenza viral protein synthesis, reproducing the in vivo situation. This selective translation is correlated with a 5- to 10-fold suppression of the dl331-induced kinase that phosphorylates the alpha subunit of eucaryotic initiation factor eIF-2. This strongly suggests that influenza virus encodes a gene product that, analogous to the adenoviral VAI RNA, prevents the shutdown of overall protein synthesis caused by an eIF-2 alpha kinase turned on by viral infection. Adenoviral mRNA translation was restored to the extract from the doubly infected cells by the addition of the guanine nucleotide exchange factor eIF-2B, which is responsible for the normal recycling of eIF-2 during protein synthesis. This indicates that the residual kinase in the doubly infected cells leads to a limitation in functional (nonsequestered) eIF-2B and hence functional (GTP-containing) eIF-2 and that under these conditions influenza viral mRNAs are selectively translated over adenoviral mRNAs. Addition of double-stranded RNA to the extracts from these cells restored the eIF-2 alpha kinase to a level approaching that seen in extracts from cells infected with dl331 alone and caused the inhibition of influenza viral mRNA translation. This suggests that the putative influenza viral gene product acts against the double-stranded RNA activation of the kinase and indicates that influenza viral mRNA translation is also linked to the level of functional eIF-2. Our results thus indicate that a limitation in functional eIF-2 which causes a nonspecific reduction in the rate of initiation of protein synthesis results in the preferential translation of the better mRNAs (influenza viral mRNAs) at the expense of the poorer mRNAs (adenoviral mRNAs).


Asunto(s)
Orthomyxoviridae/genética , Factores de Iniciación de Péptidos/genética , Biosíntesis de Proteínas , Proteínas Quinasas/genética , Procesamiento Proteico-Postraduccional , Proteínas/genética , ARN Mensajero/genética , ARN Viral/genética , Adenovirus Humanos/genética , Animales , Línea Celular , Factor 2 Eucariótico de Iniciación , Humanos , Riñón , Sustancias Macromoleculares , Mutación , Fosforilación , Conejos , Reticulocitos/metabolismo
13.
Mol Cell Biol ; 16(8): 4172-81, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8754816

RESUMEN

Expression of the double-stranded RNA-activated protein kinase (PKR) is induced by interferons, with PKR activity playing a pivotal role in establishing the interferon-induced antiviral and antiproliferative states. PKR is directly regulated by physical association with the specific inhibitor, P58IPK, a cellular protein of the tetratricopeptide repeat (TPR) family, and K3L, the product of the corresponding vaccinia virus gene. P58IPK and K3L repress PKR activation and activity. To investigate the mechanism of P58IPK- and K3L-mediated PKR inhibition, we have used a combination of in vitro and in vivo binding assays to identify the interactive regions of these proteins. The P58IPK-interacting site of PKR was mapped to a 52-amino-acid aa segment (aa 244 to 296) spanning the ATP-binding region of the protein kinase catalytic domain. The interaction with PKR did not require the C-terminal DNA-J homology region of P58IPK but was dependent on the presence of the eukaryotic initiation factor 2-alpha homology region, mapping to the 34 aa within the sixth P58IPK TPR motif. Consistent with other TPR proteins, P58IPK formed multimers in vivo: the N-terminal 166 aa were both necessary and sufficient for complex formation. A parallel in vivo analysis to map the K3L-binding region of PKR revealed that like P58IPK , K3L interacted exclusively with the PKR protein kinase catalytic domain. In contrast, however, the K3L-binding region of PKR was localized to within aa 367 to 551, demonstrating that each inhibitor bound PKR in unique, nonoverlapping domains. These data, taken together, suggest that P58IPK and K3L may mediate PKR inhibition by distinct mechanisms. Finally, we will propose a model of PKR inhibition in which P58IPK or a P58IPK complex binds PKR and interferes with nucleotide binding and autoregulation, while formation of a PKR-K3L complex interferes with active-site function and/or substrate association.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Virales/metabolismo , Secuencia de Bases , Cartilla de ADN/química , Factor 2 Eucariótico de Iniciación/química , Regulación Viral de la Expresión Génica , Sustancias Macromoleculares , Datos de Secuencia Molecular , Unión Proteica , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes , Virus Vaccinia/química , eIF-2 Quinasa
14.
Mol Cell Biol ; 18(2): 859-71, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9447982

RESUMEN

The cellular response to environmental signals is largely dependent upon the induction of responsive protein kinase signaling pathways. Within these pathways, distinct protein-protein interactions play a role in determining the specificity of the response through regulation of kinase function. The interferon-induced serine/threonine protein kinase, PKR, is activated in response to various environmental stimuli. Like many protein kinases, PKR is regulated through direct interactions with activator and inhibitory molecules, including P58IPK, a cellular PKR inhibitor. P58IPK functions to represses PKR-mediated phosphorylation of the eukaryotic initiation factor 2alpha subunit (eIF-2alpha) through a direct interaction, thereby relieving the PKR-imposed block on mRNA translation and cell growth. To further define the molecular mechanism underlying regulation of PKR, we have utilized an interaction cloning strategy to identify a novel cDNA encoding a P58IPK-interacting protein. This protein, designated P52rIPK, possesses limited homology to the charged domain of Hsp90 and is expressed in a wide range of cell lines. P52rIPK and P58IPK interacted in a yeast two-hybrid assay and were recovered as a complex from mammalian cell extracts. When coexpressed with PKR in yeast, P58IPK repressed PKR-mediated eIF-2alpha phosphorylation, inhibiting the normally toxic and growth-suppressive effects associated with PKR function. Conversely, introduction of P52rIPK into these strains resulted in restoration of both PKR activity and eIF-2alpha phosphorylation, concomitant with growth suppression due to inhibition of P58IPK function. Furthermore, P52rIPK inhibited P58IPK function in a reconstituted in vitro PKR-regulatory assay. Our results demonstrate that P58IPK is inhibited through a direct interaction with P52rIPK which, in turn, results in upregulation of PKR activity. Taken together, our data describe a novel protein kinase-regulatory system which encompasses an intersection of interferon-, stress-, and growth-regulatory pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Inhibidores Enzimáticos/metabolismo , Proteínas Represoras/metabolismo , eIF-2 Quinasa/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas Portadoras/química , Línea Celular , ADN Complementario/química , Proteínas del Choque Térmico HSP40 , Humanos , Datos de Secuencia Molecular , Unión Proteica , ARN Mensajero/metabolismo , Levaduras
15.
Mol Cell Biol ; 11(11): 5497-505, 1991 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1717830

RESUMEN

Eukaryotic viruses have devised numerous strategies to downregulate activity of the interferon-induced, double-stranded (dsRNA)-activated protein kinase (referred to as p68 on the basis of its Mr of 68,000 in human cells). Viruses must exert this control to avoid extensive phosphorylation of the alpha subunit of eukaryotic initiation factor 2 (eIF-2) by p68 and the resultant negative effects on protein synthesis initiation. To begin to define the molecular mechanisms underlying this regulation, we optimized expression of p68 in an in vitro transcription-translation system utilizing the full-length cDNA clone. The in vitro-expressed kinase was autophosphorylated in response to dsRNAs and heparin in a manner similar to that for the native p68 provided that the kinase inhibitor, 2-aminopurine, was present during the in vitro translation reaction. Further, the activated kinase efficiently phosphorylated its natural substrate, the alpha subunit of eIF-2. Binding experiments revealed that the expressed kinase complexed with the dsRNA activator, reovirus dsRNA, as well as the adenovirus-encoded inhibitor, VAI RNA. Interestingly, both the reovirus RNAs and VAI RNA also complexed with protein kinase molecules that lacked the carboxyl terminus and all catalytic domains. Deletion analysis confirmed that the p68 amino terminus contained critical determinants for reovirus dsRNA and VAI RNA binding. Further, reovirus dsRNA efficiently bound to, but failed to activate, p68 kinase molecules containing a single amino acid substitution in the invariant lysine 295 present in catalytic domain II. Taken together, these data demonstrate that this expression system permits a detailed mutagenic analysis of the regions of p68 required for interaction with virus-encoded activators and repressors.


Asunto(s)
ADN Viral/metabolismo , Interferones/farmacología , Mutagénesis Sitio-Dirigida , Biosíntesis de Proteínas , Proteínas Quinasas/genética , ARN Bicatenario/metabolismo , Adenoviridae/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sistema Libre de Células , Deleción Cromosómica , Activación Enzimática , Inducción Enzimática , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Cinética , Sustancias Macromoleculares , Datos de Secuencia Molecular , Peso Molecular , Oligonucleótidos , Fosforilación , Proteínas Quinasas/biosíntesis , Proteínas Quinasas/metabolismo , Conejos , Reoviridae/genética , Reticulocitos/metabolismo , Transcripción Genética
16.
Mol Cell Biol ; 18(9): 5208-18, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9710605

RESUMEN

The PKR protein kinase is a critical component of the cellular antiviral and antiproliferative responses induced by interferons. Recent evidence indicates that the nonstructural 5A (NS5A) protein of hepatitis C virus (HCV) can repress PKR function in vivo, possibly allowing HCV to escape the antiviral effects of interferon. NS5A presents a unique tool by which to study the molecular mechanisms of PKR regulation in that mutations within a region of NS5A, termed the interferon sensitivity-determining region (ISDR), are associated with sensitivity of HCV to the antiviral effects of interferon. In this study, we investigated the mechanisms of NS5A-mediated PKR regulation and the effect of ISDR mutations on this regulatory process. We observed that the NS5A ISDR, though necessary, was not sufficient for PKR interactions; we found that an additional 26 amino acids (aa) carboxyl to the ISDR were required for NS5A-PKR complex formation. Conversely, we localized NS5A binding to within PKR aa 244 to 296, recently recognized as a PKR dimerization domain. Consistent with this observation, we found that NS5A from interferon-resistant HCV genotype 1b disrupted kinase dimerization in vivo. NS5A-mediated disruption of PKR dimerization resulted in repression of PKR function and inhibition of PKR-mediated eIF-2alpha phosphorylation. Introduction of multiple ISDR mutations abrogated the ability of NS5A to bind to PKR in mammalian cells and to inhibit PKR in a yeast functional assay. These results indicate that mutations within the PKR-binding region of NS5A, including those within the ISDR, can disrupt the NS5A-PKR interaction, possibly rendering HCV sensitive to the antiviral effects of interferon. We propose a model of PKR regulation by NS5A which may have implications for therapeutic strategies against HCV.


Asunto(s)
Hepacivirus/fisiología , Interferones/farmacología , Proteínas no Estructurales Virales/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Células COS , Clonación Molecular , Cartilla de ADN , Dimerización , Escherichia coli , Regulación Enzimológica de la Expresión Génica , Hepacivirus/efectos de los fármacos , Hepacivirus/patogenicidad , Modelos Biológicos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación Puntual , Reacción en Cadena de la Polimerasa , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Eliminación de Secuencia , Transfección , Proteínas no Estructurales Virales/biosíntesis , Proteínas no Estructurales Virales/química , Replicación Viral , eIF-2 Quinasa/química
17.
Mol Cell Biol ; 18(12): 7304-16, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9819417

RESUMEN

The human double-stranded RNA (dsRNA)-dependent protein kinase PKR inhibits protein synthesis by phosphorylating translation initiation factor 2alpha (eIF2alpha). Vaccinia virus E3L encodes a dsRNA binding protein that inhibits PKR in virus-infected cells, presumably by sequestering dsRNA activators. Expression of PKR in Saccharomyces cerevisiae inhibits protein synthesis by phosphorylation of eIF2alpha, dependent on its two dsRNA binding motifs (DRBMs). We found that expression of E3 in yeast overcomes the lethal effect of PKR in a manner requiring key residues (Lys-167 and Arg-168) needed for dsRNA binding by E3 in vitro. Unexpectedly, the N-terminal half of E3, and residue Trp-66 in particular, also is required for anti-PKR function. Because the E3 N-terminal region does not contribute to dsRNA binding in vitro, it appears that sequestering dsRNA is not the sole function of E3 needed for inhibition of PKR. This conclusion was supported by the fact that E3 activity was antagonized, not augmented, by overexpressing the catalytically defective PKR-K296R protein containing functional DRBMs. Coimmunoprecipitation experiments showed that a majority of PKR in yeast extracts was in a complex with E3, whose formation was completely dependent on the dsRNA binding activity of E3 and enhanced by the N-terminal half of E3. In yeast two-hybrid assays and in vitro protein binding experiments, segments of E3 and PKR containing their respective DRBMs interacted in a manner requiring E3 residues Lys-167 and Arg-168. We also detected interactions between PKR and the N-terminal half of E3 in the yeast two-hybrid and lambda repressor dimerization assays. In the latter case, the N-terminal half of E3 interacted with the kinase domain of PKR, dependent on E3 residue Trp-66. We propose that effective inhibition of PKR in yeast requires formation of an E3-PKR-dsRNA complex, in which the N-terminal half of E3 physically interacts with the protein kinase domain of PKR.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteínas de Unión al ARN/genética , Proteínas Virales/genética , eIF-2 Quinasa/genética , División Celular/genética , Dimerización , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica/genética , Mutación/genética , Fosforilación , Unión Proteica/genética , Proteínas Recombinantes de Fusión/genética , Ribosomas/genética , Saccharomyces cerevisiae/genética
18.
Mol Cell Biol ; 19(7): 4757-65, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10373525

RESUMEN

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.


Asunto(s)
Apoptosis , Chaperonas Moleculares/metabolismo , Inhibidores de Proteínas Quinasas , ARN Bicatenario/metabolismo , Proteínas Represoras/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , eIF-2 Quinasa/metabolismo , Células 3T3 , Animales , Factor 2 Eucariótico de Iniciación/metabolismo , Proteínas del Choque Térmico HSP40 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Chaperonas Moleculares/genética , Mutagénesis , FN-kappa B/metabolismo , Fenotipo , Fosforilación , Poli I-C/metabolismo , Poli I-C/farmacología , ARN Bicatenario/antagonistas & inhibidores , Conejos , Proteínas Represoras/genética , Transformación Genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología
19.
Trends Microbiol ; 3(2): 75-8, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7537157

RESUMEN

Viruses that fail to block the lethal effects of the double-stranded-RNA-activated protein kinase (PKR) may be doomed; why do so many viruses go to so much trouble to downregulate this interferon-induced protein kinase? PKR may regulate cell growth and proliferation in uninfected cells, suggesting that it also participates in the antiproliferative arm of the interferon response.


Asunto(s)
Regulación Viral de la Expresión Génica , Interferones/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Fenómenos Fisiológicos de los Virus , Animales , Transformación Celular Neoplásica , Inducción Enzimática , Proteínas del Choque Térmico HSP40 , Humanos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Represoras/fisiología , eIF-2 Quinasa
20.
Pharmacol Ther ; 78(1): 29-46, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9593328

RESUMEN

The interferon (IFN)-induced cellular antiviral response is the first line of defense against viral infection within an animal host. In order to establish a productive infection, eukaryotic viruses must first overcome the IFN-induced blocks imposed on viral replication. The double-stranded RNA-activated protein kinase (PKR) is a key component mediating the antiviral actions of IFN. This IFN-induced protein kinase can restrict viral replication through its ability to phosphorylate the protein synthesis initiation factor eukaryotic initiation factor-2 alpha-subunit and reduce levels of viral protein synthesis. Viruses, therefore, must block the function of PKR in order to avoid these deleterious antiviral effects associated with PKR activity. Indeed, many viruses have developed effective measures to repress PKR activity during infection. This review will focus primarily on an overview of the different molecular mechanisms employed by these viruses to meet a common goal: the inhibition of PKR function, uncompromised viral protein synthesis, and unrestricted virus replication. The past few years have seen exciting new advances in this area. Rather unexpectedly, this area of research has benefited from the use of the yeast system to study PKR. Other recent advances include studies on PKR regulation by the herpes simplex viruses and data from our laboratory on the medically important hepatitis C viruses. We speculate that IFN is ineffective as a therapeutic agent against hepatitis C virus because the virus can effectively repress PKR function. Finally, we will discuss briefly the future directions of this PKR field.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Microbiana , Interferones/farmacología , Proteínas Virales/fisiología , Replicación Viral/efectos de los fármacos , eIF-2 Quinasa/antagonistas & inhibidores , Animales , Antivirales/uso terapéutico , Humanos , Interferones/uso terapéutico , Biosíntesis de Proteínas/efectos de los fármacos , Virosis/tratamiento farmacológico , Virosis/virología , eIF-2 Quinasa/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda