Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Virol ; 85(1): 286-95, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21047956

RESUMEN

The human adenovirus E4orf4 protein is toxic in both human tumor cells and Saccharomyces cerevisiae. Previous studies indicated that most of this toxicity is dependent on an interaction of E4orf4 protein with the B55 class of regulatory subunits of protein phosphatase 2A (PP2A) and in yeast with the B55 homolog Cdc55. We have found previously that E4orf4 inhibits PP2A activity against at least some substrates. In an attempt to understand the mechanism of this inhibition, we used a genetic approach to identify residues in the seven-bladed ß-propeller proteins B55α and Cdc55 required for E4orf4 binding. In both cases, amino-terminal polypeptides composed only of blade 1 and at least part of blade 2 were found to bind E4orf4 and overexpression blocked E4orf4 toxicity in yeast. Furthermore, certain amino acid substitutions in blades 1 and 2 within full-length B55α and Cdc55 resulted in loss of E4orf4 binding. Recent mutational analysis has suggested that segments of blades 1 and 2 present on the top face of B55α form part of the "substrate-binding groove." Additionally, these segments are in close proximity to the catalytic C subunit of the PP2A holoenzyme. Thus, our results are consistent with the hypothesis that E4orf4 binding could affect the access of substrates, resulting in the failure to dephosphorylate some PP2A substrates.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteína Fosfatasa 2/genética , Subunidades de Proteína/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas Virales/metabolismo , Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Secuencia de Aminoácidos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/metabolismo , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Técnicas del Sistema de Dos Híbridos , Proteínas Virales/genética
2.
J Virol ; 83(4): 1689-99, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19073741

RESUMEN

The human adenovirus type 5 (Ad5) E4orf4 product has been studied extensively although in most cases as expressed from vectors in the absence of other viral products. Thus, relatively little is known about its role in the context of an adenovirus infection. Although considerable earlier work had indicated that the E4orf4 protein is not essential for replication, a recent study using dl359, an Ad5 mutant believed to produce a nonfunctional E4orf4 protein, suggested that E4orf4 is essential for virus growth in primary small-airway epithelial cells (C. O'Shea, et al., EMBO J. 24:1211-1221, 2005). Hence, to examine further the role of E4orf4 during virus infection, we generated for the first time a set of E4orf4 virus mutants in a common Ad5 genetic background. Such mutant viruses included those that express E4orf4 proteins containing various individual point mutations, those defective entirely in E4orf4 expression, and a mutant expressing wild-type E4orf4 fused to the green fluorescent protein. E4orf4 protein was found to localize primarily in nuclear structures shown to be viral replication centers, in nucleoli, and in perinuclear bodies. Importantly, E4orf4 was shown not to be essential for virus growth in either human tumor or primary cells, at least in tissue culture. Unlike E4orf4-null virus, mutant dl359 appeared to exhibit a gain-of-function phenotype that impairs virus growth. The dl359 E4orf4 protein, which contains a large in-frame internal deletion, clustered in aggregates enriched in Hsp70 and proteasome components. In addition, the late viral mRNAs produced by dl359 accumulated abnormally in a nuclear punctate pattern. Altogether, our results indicate that E4orf4 protein is not essential for virus growth in culture and that expression of the dl359 E4orf4 product interferes with viral replication, presumably through interactions with structures in the nucleus.


Asunto(s)
Adenovirus Humanos/fisiología , Proteínas Virales/metabolismo , Replicación Viral , Línea Celular , Nucléolo Celular/química , Núcleo Celular/química , Citoplasma/química , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Virales/genética
3.
J Virol ; 83(17): 8340-52, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19535438

RESUMEN

Human adenovirus E4orf4 protein is toxic in human tumor cells. Its interaction with the B alpha subunit of protein phosphatase 2A (PP2A) is critical for cell killing; however, the effect of E4orf4 binding is not known. B alpha is one of several mammalian B-type regulatory subunits that form PP2A holoenzymes with A and C subunits. Here we show that E4orf4 protein interacts uniquely with B55 family subunits and that cell killing increases with the level of E4orf4 expression. Evidence suggesting that B alpha-specific PP2A activity, measured in vitro against phosphoprotein substrates, is reduced by E4orf4 binding was obtained, and two potential B55-specific PP2A substrates, 4E-BP1 and p70(S6K), were seen to be hypophosphorylated in vivo following expression of E4orf4. Furthermore, treatment of cells with low levels of the phosphatase inhibitor okadaic acid or coexpression of the PP2A inhibitor I(1)(PP2A) enhanced E4orf4-induced cell killing and G(2)/M arrest significantly. These results suggested that E4orf4 toxicity results from the inhibition of B55-specific PP2A holoenzymes, an idea that was strengthened by an observed growth arrest resulting from treatment of H1299 cells with B alpha-specific RNA interference. We believe that E4orf4 induces growth arrest resulting in cell death by reducing the global level of B55-specific PP2A activity, thus preventing the dephosphorylation of B55-specific PP2A substrates, including those involved in cell cycle progression.


Asunto(s)
Adenovirus Humanos/patogenicidad , Ciclo Celular , Muerte Celular , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteínas Virales/metabolismo , Recuento de Células , Línea Celular Tumoral , Supervivencia Celular , Humanos , Unión Proteica , Proteína Fosfatasa 2/metabolismo
4.
Mol Cell Biol ; 24(8): 3415-29, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15060162

RESUMEN

Phosphorylation of the alpha subunit of eukaryotic translation initiation factor 2 (eIF2alpha) at serine 51 inhibits protein synthesis in cells subjected to various forms of stress including virus infection. The human papillomavirus (HPV) E6 oncoprotein contributes to virus-induced pathogenicity through multiple mechanisms including the inhibition of apoptosis and the blockade of interferon (IFN) action. We have investigated a possible functional relationship between the E6 oncoprotein and eIF2alpha phosphorylation by an inducible-dimerization form of the IFN-inducible protein kinase PKR. Herein, we demonstrate that HPV type 18 E6 protein synthesis is rapidly repressed upon eIF2alpha phosphorylation caused by the conditional activation of the kinase. The remainder of E6, however, can rescue cells from PKR-mediated inhibition of protein synthesis and induction of apoptosis. E6 physically associates with GADD34/PP1 holophosphatase complex, which mediates translational recovery, and facilitates eIF2alpha dephosphorylation. Inhibition of eIF2alpha phosphorylation by E6 mitigates eIF2alpha-dependent responses to transcription and translation of proapoptotic genes. These findings demonstrate, for the first time, a role of the oncogenic E6 in apoptotic signaling induced by PKR and eIF2alpha phosphorylation. The functional interaction between E6 and the eIF2alpha phosphorylation pathway may have important implications for HPV infection and associated pathogenesis.


Asunto(s)
Muerte Celular/fisiología , Proteínas de Unión al ADN , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica , Proteínas Oncogénicas Virales/metabolismo , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2 , Aminocumarinas , Antígenos de Diferenciación , Proteínas de Ciclo Celular , Línea Celular Tumoral , Cumarinas/metabolismo , Activación Enzimática , Factor 2 Eucariótico de Iniciación/química , Factor 2 Eucariótico de Iniciación/genética , Humanos , Interferón-alfa/metabolismo , Proteínas Oncogénicas Virales/genética , Papillomaviridae/metabolismo , Infecciones por Papillomavirus , Fosforilación , Biosíntesis de Proteínas , Proteína Fosfatasa 1 , Subunidades de Proteína/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , Transcripción Genética , Proteína X Asociada a bcl-2 , eIF-2 Quinasa/metabolismo
5.
Cancer Res ; 70(20): 7820-9, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20924113

RESUMEN

Hypoxia within the tumor microenvironment promotes angiogenesis, metabolic reprogramming, and tumor progression. In addition to activating hypoxia-inducible factor-1α (HIF-1α), cells also respond to hypoxia by globally inhibiting protein synthesis via serine 51 phosphorylation of translation eukaryotic initiation factor 2α (eIF2α). In this study, we investigated potential roles for stress-activated eIF2α kinases in regulation of HIF-1α. Our investigations revealed that the double-stranded RNA-dependent protein kinase R (PKR) plays a significant role in suppressing HIF-1α expression, acting specifically at the level of transcription. HIF-1α transcriptional repression by PKR was sufficient to impair the hypoxia-induced accumulation of HIF-1α and transcriptional induction of HIF-1α-dependent target genes. Inhibition of HIF-1A transcription by PKR was independent of eIF2α phosphorylation but dependent on inhibition of the signal transducer and activator of transcription 3 (Stat3). Furthermore, HIF-1A repression required the T-cell protein tyrosine phosphatase, which acts downstream of PKR, to suppress Stat3. Our findings reveal a novel tumor suppressor function for PKR, which inhibits HIF-1α expression through Stat3 but is independent of eIF2α phosphorylation.


Asunto(s)
Hipoxia de la Célula/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transcripción Genética , eIF-2 Quinasa/metabolismo , Actinas/genética , Animales , Genes Reporteros , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Noqueados , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Supresión Genética , Factor A de Crecimiento Endotelial Vascular/genética , eIF-2 Quinasa/genética
6.
EMBO Rep ; 8(3): 265-70, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17290288

RESUMEN

The interferon (IFN)-inducible, double-stranded RNA activated protein kinase (PKR) is a dual-specificity kinase, which has an essential role in the regulation of protein synthesis by phosphorylating the translation eukaryotic initiation factor 2 (eIF2). Here, we show the tyrosine (Tyr) phosphorylation of PKR in response to type I or type II IFNs. We show that PKR physically interacts with either Jak1 or Tyk2 in unstimulated cells and that these interactions are increased in IFN-treated cells. We also show that PKR acts as a substrate of activated Jaks, and is phosphorylated at Tyr 101 and Tyr 293 both in vitro and in vivo. Moreover, we provide strong evidence that both the induction of eIF2alpha phosphorylation and inhibition of protein synthesis by IFN are impaired in cells lacking Jak1 or Tyk2, which corresponds to a lack of induction of PKR tyrosine phosphorylation. We conclude that PKR tyrosine phosphorylation provides an important link between IFN signalling and translational control through the regulation of eIF2alpha phosphorylation.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Interferones/metabolismo , Janus Quinasa 1/metabolismo , Transducción de Señal/fisiología , TYK2 Quinasa/metabolismo , Tirosina/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Línea Celular , Electroforesis en Gel de Poliacrilamida , Immunoblotting , Ratones , Fosforilación
7.
J Biol Chem ; 277(41): 38364-72, 2002 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-12161430

RESUMEN

The interferon-inducible double-stranded RNA (dsRNA)-activated protein kinase, PKR, plays an important role in messenger (m) RNA translation by phosphorylating the alpha subunit of eukaryotic initiation factor 2. Through this capacity PKR is thought to be a mediator of the antiviral and antiproliferative actions of interferon. In addition to translational function, PKR has been implicated in many signaling pathways to gene transcription by modulating the activities of a number of transcription factors, including NF-kappa B and STATs. However, experiments with two different PKR knockout (PKR(-/-)) mouse models have failed to verify many of the biological functions attributed to PKR. In addition, results with cells from the two PKR(-/-) mice have been contradictory and confusing. Here, we show that the first PKR(-/-) mouse with deletion of exons 2 and 3, corresponding to the N terminus domain of PKR (N-PKR(-/-)), expresses a truncated protein, resulting from the translation of the exon-skipped mouse PKR (ES-mPKR) mRNA. The ES-mPKR protein is defective in dsRNA binding but remains catalytically active both in vitro and in vivo. Furthermore, we show that the second PKR(-/-) mouse with a targeted deletion of exon 12, which corresponds to the C terminus of the molecule (C-PKR(-/-)), expresses a truncated mPKR produced by alternative splicing of exon 12. Although the spliced form of mPKR (SF-mPKR) is catalytically inactive, it retains the dsRNA-binding properties of the wild type mPKR. Reverse transcription-PCRs demonstrate that SF-mPKR mRNA is expressed in several normal mouse tissues, and appears to be under developmental control during embryogenesis. Our data demonstrate that both PKR(-/-) models are incomplete knockouts, and expression of the PKR variants may account, at least in part, for the significant signaling differences between cells from the two PKR(-/-) mice.


Asunto(s)
Proteínas I-kappa B , Isoenzimas/metabolismo , Mutación , eIF-2 Quinasa/metabolismo , Empalme Alternativo , Animales , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Células HeLa , Humanos , Isoenzimas/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidor NF-kappaB alfa , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , ARN Bicatenario/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/fisiología , Distribución Tisular , eIF-2 Quinasa/genética
8.
J Gen Virol ; 85(Pt 6): 1419-1426, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15166424

RESUMEN

Human papillomaviruses (HPVs) are aetiological agents for genital warts and cervical cancer, the different pathologies of which are dependent on the type of HPV infection. Oncogenic HPV types associated with cancer are carcinogens by virtue of their oncogene products, which target key regulators of cell proliferation and apoptosis. The viral E6 protein from oncogenic HPV types plays a central role in carcinogenesis by exploiting the cellular proteasome degradation pathway in order to mediate the degradation of cellular proteins, most notably the prototype tumour suppressor protein p53. Much less is known about the cellular targets of E6 from the non-oncogenic HPV types associated with genital warts. It is also unclear what factors influence the level and stability of the viral E6 proteins in cells. This report demonstrates that both oncogenic and non-oncogenic HPV E6 proteins (from types 18 and 11, respectively) are ubiquitinated and targeted for degradation by the 26S proteasome. E6 domains required for the induction of p53 or DLG degradation, or E6AP binding, are not involved in proteasome-mediated degradation of HPV-18 E6. These results provide insight into the cellular modulation of E6 protein levels from both high-risk and low-risk HPV types.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Proteínas de Unión al ADN , Complejos Multienzimáticos/fisiología , Proteínas Oncogénicas Virales/metabolismo , Ubiquitina/metabolismo , Línea Celular , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/metabolismo , Complejo de la Endopetidasa Proteasomal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA