Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Virol ; 85(18): 9447-58, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21734036

RESUMEN

In recent studies, the nuclear domain 10 (ND10) components PML and hDaxx were identified as cellular restriction factors that inhibit the initiation of human cytomegalovirus (HCMV) replication. The antiviral function of ND10, however, is antagonized by the IE1 protein, which induces ND10 disruption. Here we show that IE1 not only de-SUMOylates PML immediately upon infection but also directly targets Sp100. IE1 expression alone was sufficient to downregulate endogenous Sp100 independently of the presence of PML. Moreover, cotransfection experiments revealed that IE1 negatively interferes with the SUMOylation of all Sp100 isoforms. The modulation of Sp100 at immediate-early (IE) times of infection, indeed, seemed to have an in vivo relevance for HCMV replication, since knockdown of Sp100 resulted in more cells initiating the viral gene expression program. In addition, we observed that Sp100 was degraded in a proteasome-dependent manner at late times postinfection, suggesting that Sp100 may play an additional antiviral role during the late phase. Infection experiments conducted with Sp100 knockdown human foreskin fibroblasts (HFFs) confirmed this hypothesis: depletion of Sp100 resulted in augmented release of progeny virus particles compared to that from control cells. Consistent with this observation, we noted increased amounts of viral late gene products in the absence of Sp100. Importantly, this elevated late gene expression was not dependent on enhanced viral IE gene expression. Taken together, our data provide evidence that Sp100 is the first ND10-related factor identified that not only possesses the potential to restrict the initial stage of infection but also inhibits HCMV replication during the late phase.


Asunto(s)
Antígenos Nucleares/inmunología , Autoantígenos/inmunología , Citomegalovirus/inmunología , Citomegalovirus/fisiología , Interacciones Huésped-Patógeno , Replicación Viral , Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Línea Celular , Fibroblastos/inmunología , Fibroblastos/virología , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Sumoilación
2.
J Gen Virol ; 92(Pt 7): 1532-1538, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21471311

RESUMEN

Nuclear domains 10 (ND10s) are discrete subnuclear structures that contain the three major protein components promyelocytic leukaemia protein (PML), hDaxx and Sp100. Previous studies identified the ND10-components PML and hDaxx as cellular restriction factors that independently counteract human cytomegalovirus (HCMV) infection via the repression of viral immediate-early (IE) gene expression. Consequently, we asked whether Sp100 is likewise involved in this repressive activity. Infection of Sp100 knockdown (kd) cells with HCMV resulted in a significantly increased plaque-forming ability. In addition, ablation of Sp100 led to a considerable increase in the number of IE1-expressing cells, indicating that Sp100 suppresses the initiation of viral gene expression. Next, double-kd cells, lacking either Sp100/hDaxx or Sp100/PML, were generated. Here, infection resulted in an additional enhancement in HCMV replication efficacy compared with the single-kd cells. Thus, our results further strengthen the concept that the three major ND10-components independently contribute to the cellular restriction of HCMV replication.


Asunto(s)
Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/genética , Regulación Viral de la Expresión Génica , Proteínas Inmediatas-Precoces/genética , Proteínas Nucleares/metabolismo , Antígenos Nucleares/genética , Autoantígenos/genética , Línea Celular , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Nucleares/genética , Replicación Viral
3.
J Gen Virol ; 92(Pt 12): 2757-2769, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21832009

RESUMEN

The onset of human cytomegalovirus (HCMV) lytic replication is strictly controlled by the host cell division cycle. Although viral entry of S/G2-phase cells is unperturbed expression of major immediate-early (MIE) genes IE1 and IE2 is tightly blocked in these cells. Besides the finding that cyclin-dependent kinase (CDK) activity is required for IE1/IE2 repression little is known about the nature of this cell cycle-dependent block. Here, we show that the block occurs after nuclear entry of viral DNA and prevents the accumulation of IE1/IE2 mRNAs, suggesting an inhibition of transcription. Remarkably, the presence of cis-regulatory regions of the MIE locus is neither sufficient nor necessary for IE1/IE2 repression in the S/G2 phase. Furthermore, the block of viral mRNA expression also affects other immediate-early transcribed regions, i.e. the US3 and UL36-38 gene loci. This suggests a mechanism of repression that acts in a general and not a gene-specific fashion. Such a nuclear, genome-wide repression of HCMV is typically mediated by the intrinsic immune defence at nuclear domain 10 (ND10) structures. However, we found that neither Daxx nor PML, the main players of ND10-based immunity, are required for the block to viral gene expression in the S/G2 phase. In addition, the viral tegument protein pp71 (pUL82), a major antagonist of the intrinsic immunity at pre-immediate-early times of infection, proved to be functional in S-phase cells. This suggests the existence of a yet undiscovered, CDK-dependent mechanism exerting higher-level control over immediate-early mRNA expression in HCMV-infected cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Citomegalovirus/genética , Regulación Viral de la Expresión Génica , Genes Inmediatos-Precoces , Proteínas Nucleares/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Células 3T3 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Clonación Molecular , Proteínas Co-Represoras , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Citomegalovirus/metabolismo , Citomegalovirus/fisiología , ADN Viral/genética , ADN Viral/metabolismo , Fase G2 , Sitios Genéticos , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Ratones , Chaperonas Moleculares , Mutagénesis , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fase S , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Virales/metabolismo , Replicación Viral
4.
J Virol ; 83(24): 12881-94, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19812159

RESUMEN

The major transactivator protein IE2p86 of human cytomegalovirus (HCMV) has previously been shown to undergo posttranslational modification by the covalent attachment of SUMO proteins, termed SUMOylation, which occurs at two lysine residues located at amino acid positions 175 and 180. Mutation of the acceptor lysines resulted in the abrogation of IE2p86 SUMOylation in mammalian cells and a strong reduction of IE2p86-mediated transactivation. In this paper, we identify an additional SUMO interaction motif (SIM) within IE2p86, which mediates noncovalent binding to SUMO, as shown by yeast two-hybrid analyses. Transient-expression experiments revealed that an IE2p86 SIM mutant exhibited significantly reduced SUMOylation, strongly suggesting that noncovalent SUMO interactions affect the efficacy of covalent SUMO coupling. In order to define the relevance of IE2p86 SUMO interactions for viral replication, recombinant viruses originating from two different HCMV strains (AD169 and VR1814) were generated. Analysis of viruses expressing SUMOylation-negative IE2p86 revealed strongly impaired replication due to reduced viral DNA and protein accumulation, as well as diminished initiation of immediate-early gene expression. The additional introduction of the SIM mutation into the viral genome did not further compromise viral replication but resulted in altered expression of viral proteins at late times postinfection. In summary, this paper clearly shows that IE2p86 SUMOylation is necessary for efficient replication of the HCMV laboratory strain AD169 and the clinical isolate VR1814 and thus for the in vivo function of this viral transcription factor.


Asunto(s)
Citomegalovirus/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Transactivadores/metabolismo , Células Cultivadas , ADN Viral/metabolismo , Exones , Humanos , Proteínas Inmediatas-Precoces/genética , Transactivadores/genética , Replicación Viral
5.
Biochim Biophys Acta ; 1783(11): 2207-21, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18775455

RESUMEN

Nuclear domains 10 (ND10), alternatively termed PML nuclear bodies (PML-NBs) or PML oncogenic domains (PODs), have been discovered approximately 15 years ago as a nuclear substructure that is targeted by a variety of viruses belonging to different viral families. This review will summarize the most important structural and functional characteristics of ND10 and its major protein constituents followed by a discussion of the current view regarding the role of this subnuclear structure for various DNA and RNA viruses with an emphasis on herpesviruses. It is concluded that accumulating evidence argues for an involvement of ND10 in host antiviral defenses either via mediating an intrinsic immune response against specific viruses or via acting as a component of the cellular interferon pathway.


Asunto(s)
Núcleo Celular , Cuerpos de Inclusión Intranucleares , Proteínas Nucleares/metabolismo , Virosis , Virus , Animales , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Núcleo Celular/virología , Humanos , Cuerpos de Inclusión Intranucleares/metabolismo , Cuerpos de Inclusión Intranucleares/virología , Proteínas Nucleares/genética , Virosis/metabolismo , Virosis/patología , Replicación Viral , Virus/genética , Virus/metabolismo
6.
J Virol ; 82(1): 126-37, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17942542

RESUMEN

Infection with DNA viruses commonly results in the association of viral genomes with a cellular subnuclear structure known as nuclear domain 10 (ND10). Recent studies demonstrated that individual ND10 components, like hDaxx or promyelocytic leukemia protein (PML), mediate an intrinsic immune response against human cytomegalovirus (HCMV) infection, strengthening the assumption that ND10 components are part of a cellular antiviral defense mechanism. In order to further define the role of hDaxx and PML for HCMV replication, we generated either primary human fibroblasts with a stable, individual knockdown of PML or hDaxx (PML-kd and hDaxx-kd, respectively) or cells exhibiting a double knockdown. Comparative analysis of HCMV replication in PML-kd or hDaxx-kd cells revealed that immediate-early (IE) gene expression increased to a similar extent, regardless of which ND10 constituent was depleted. Since a loss of PML, the defining component of ND10, results in a dispersal of the entire nuclear substructure, the increased replication efficacy of HCMV in PML-kd cells could be a consequence of the dissociation of the repressor protein hDaxx from its optimal subnuclear localization. However, experiments using three different recombinant HCMVs revealed a differential growth complementation in PML-kd versus hDaxx-kd cells, strongly arguing for an independent involvement in suppressing HCMV replication. Furthermore, infection experiments using double-knockdown cells devoid of both PML and hDaxx illustrated an additional enhancement in the replication efficacy of HCMV compared to the single-knockdown cells. Taken together, our data indicate that both proteins, PML and hDaxx, mediate an intrinsic immune response against HCMV infection by contributing independently to the silencing of HCMV IE gene expression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Proteínas de Neoplasias/fisiología , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología , Replicación Viral , Células Cultivadas , Proteínas Co-Represoras , Citomegalovirus/genética , Fibroblastos/virología , Silenciador del Gen , Humanos , Chaperonas Moleculares , Proteínas de Neoplasias/química , Proteínas Nucleares/química , Proteína de la Leucemia Promielocítica , Estructura Terciaria de Proteína , Factores de Transcripción/química , Proteínas Supresoras de Tumor/química
7.
J Virol ; 82(21): 10543-55, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18715911

RESUMEN

The human cytomegalovirus (HCMV) UL82-encoded tegument protein pp71 has recently been shown to activate viral immediate-early (IE) gene expression by neutralizing a cellular intrinsic immune defense instituted by the ND10 protein hDaxx. Pp71 localizes to ND10 upon infection and induces the degradation of hDaxx. Here, we report the successful generation of a recombinant HCMV expressing enhanced yellow fluorescent protein (EYFP) fused to the N terminus of pp71. Intriguingly, insertion of the EYFP-UL82 coding sequence into the HCMV AD169 genome gave rise to a recombinant virus, termed AD169/EYFP-pp71, that replicates to significantly higher titers than wild-type AD169. In particular, we noticed strongly increased protein levels of pp71 after AD169/EYFP-pp71 inoculation. Although the high abundance of pp71 resulted in augmented packaging of the tegument protein into viral particles, no increased hDaxx degradation was detectable upon AD169/EYFP-pp71 infection. In contrast, further investigation revealed a significantly enhanced viral DNA replication compared to wild-type AD169. Thus, we hypothesize that an as-yet-unidentified function of pp71 contributes to the enhanced infectivity of AD169/EYFP-pp71. This assumption is additionally supported by the observation that increased early and late gene expression after AD169/EYFP-pp71 infection occurs independent of elevated IE protein levels. Finally, immunofluorescence analyses confirmed that hDaxx determines the ND10-localization of pp71 upon infection, since pp71 exhibited a nucleolar distribution in the absence of hDaxx. Taken together, we generated a recombinant HCMV that constitutes a useful tool not only to dissect the in vivo dynamics of pp71 subnuclear localization more precisely but also to explore new features of this viral transactivator.


Asunto(s)
Proteínas Bacterianas/genética , Citomegalovirus/crecimiento & desarrollo , Citomegalovirus/genética , Proteínas Luminiscentes/genética , Mutagénesis Insercional , Proteínas Recombinantes de Fusión/genética , Proteínas Virales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Cultivadas , Proteínas Co-Represoras , Replicación del ADN , ADN Viral/biosíntesis , Humanos , Chaperonas Moleculares , Proteínas Nucleares/metabolismo , Proteínas Virales/biosíntesis , Replicación Viral
8.
FEBS Lett ; 582(5): 666-72, 2008 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-18242185

RESUMEN

This study provides evidence that proteasomal activity is required at multiple steps in human cytomegalovirus replication. Electron microscopy revealed that no viral particles were assembled in the presence of proteasome inhibitor MG132. Immunofluorescence and Western blot analyses using MG132 demonstrated that immediate early gene expression was suppressed at low but not high MOI. In contrast, expression of late proteins was completely blocked independent of MOI. Additionally, pulsed-field gel electrophoresis demonstrated that MG132 interferes with cleavage of HCMV DNA. Bromodeoxyuridine incorporation studies showed that de novo viral DNA synthesis is reduced in the presence of MG132. Furthermore, in contrast to previous hypotheses we demonstrated that neither the ND10 components PML and hDaxx nor NFkappaB activation represent the target for MG132.


Asunto(s)
Citomegalovirus/efectos de los fármacos , Citomegalovirus/fisiología , ADN Viral/biosíntesis , Leupeptinas/farmacología , Inhibidores de Proteasoma , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Antivirales/farmacología , Células Cultivadas , Infecciones por Citomegalovirus/virología , ADN Concatenado/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Humanos , FN-kappa B/deficiencia , FN-kappa B/metabolismo , Proteínas Virales/metabolismo , Virión/efectos de los fármacos , Virión/metabolismo
9.
Virus Res ; 157(2): 128-33, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20934469

RESUMEN

In recent studies we and others have identified the cellular proteins PML, hDaxx, Sp100 and ATRX, which form a subnuclear structure known as nuclear domain 10 (ND10) or PML nuclear bodies (PML-NBs), as host restriction factors that counteract cytomegalovirus infections by inhibiting the initiation of viral immediate-early (IE) gene expression. The antiviral function of ND10, however, is antagonized by viral regulatory proteins which either induce a proteasomal degradation of ND10 proteins or a disruption of the subnuclear structure. This review will summarize our current knowledge on the inhibition of cytomegalovirus replication by ND10 proteins. Furthermore, viral strategies to defeat this host defense mechanism are discussed.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Nucleares/inmunología , Citomegalovirus/patogenicidad , Citomegalovirus/fisiología , Regulación Viral de la Expresión Génica , Humanos , Inmunidad Innata , Proteínas Virales/metabolismo , Latencia del Virus , Replicación Viral
10.
Viruses ; 1(3): 1240-64, 2009 12.
Artículo en Inglés | MEDLINE | ID: mdl-21994592

RESUMEN

In recent studies we and others have identified the cellular proteins PML, hDaxx, and Sp100, which form a subnuclear structure known as nuclear domain 10 (ND10) or PML nuclear bodies (PML-NBs), as host restriction factors that counteract herpesviral infections by inhibiting viral replication at different stages. The antiviral function of ND10, however, is antagonized by viral regulatory proteins (e.g., ICP0 of herpes simplex virus; IE1 of human cytomegalovirus) which induce either a modification or disruption of ND10. This review will summarize the current knowledge on how viral replication is inhibited by ND10 proteins. Furthermore, herpesviral strategies to defeat this host defense mechanism are discussed.

11.
J Virol ; 81(18): 10123-36, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17626080

RESUMEN

The human cytomegalovirus (HCMV) immediate-early 2 (IE2) transactivator has previously been shown to form intranuclear, dot-like accumulations in association with subnuclear structures known as promyelocytic leukemia protein (PML) nuclear bodies or ND10. We recently observed that IE2 can form dot-like structures even after infection of PML knockdown cells, which lack genuine ND10. To further analyze the determinants of IE2 subnuclear localization, a recombinant HCMV expressing IE2 fused to the enhanced green fluorescent protein was constructed. We infected primary human fibroblasts expressing Sp100 fused to the autofluorescent protein mCherry while performing live-cell imaging experiments. These experiments revealed a very dynamic association of IE2 dots with ND10 structures during the first hours postinfection: juxtaposed structures rapidly fused to precise co-localizations, followed by segregation, and finally, the dispersal of ND10 accumulations. Furthermore, by infecting PML knockdown cells we determined that the number of IE2 accumulations was dependent on the multiplicity of infection. Since time-lapse microscopy in live-infected cells revealed that IE2 foci developed into viral replication compartments, we hypothesized that viral DNA could act as a determinant of IE2 accumulations. Direct evidence that IE2 molecules are associated with viral DNA early after HCMV infection was obtained using fluorescence in situ hybridization. Finally, a DNA-binding-deficient IE2 mutant could no longer be recruited into viral replication centers, suggesting that the association of IE2 with viral DNA is mediated by a direct DNA contact. Thus, we identified viral DNA as an important determinant of IE2 subnuclear localization, which suggests that the formation of a virus-induced nucleoprotein complex and its spatial organization is likely to be critical at the early stages of a lytic infection.


Asunto(s)
Núcleo Celular/metabolismo , Citomegalovirus/metabolismo , Genoma Viral , Proteínas Inmediatas-Precoces/metabolismo , Cuerpos de Inclusión Viral/metabolismo , Cuerpos de Inclusión Intranucleares/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Transporte Activo de Núcleo Celular/genética , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Línea Celular , Núcleo Celular/genética , Núcleo Celular/patología , Núcleo Celular/virología , Citomegalovirus/genética , ADN Viral/genética , ADN Viral/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/virología , Genoma Viral/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Cuerpos de Inclusión Viral/genética , Cuerpos de Inclusión Viral/virología , Cuerpos de Inclusión Intranucleares/genética , Cuerpos de Inclusión Intranucleares/virología , Mutación , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteína de la Leucemia Promielocítica , Transactivadores/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Replicación Viral/genética
12.
J Virol ; 80(16): 8006-18, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16873257

RESUMEN

Several viruses, including human cytomegalovirus (HCMV), encode proteins that colocalize with a cellular subnuclear structure known as ND10. Since only viral DNA deposited at ND10 initiates transcription, ND10 structures were hypothesized to be essential for viral replication. On the other hand, interferon treatment induces an up-regulation of ND10 structures and viruses have evolved polypeptides that disperse the dot-like accumulation of ND10 proteins, suggesting that ND10 could also be part of an intrinsic defense mechanism. In order to obtain evidence for either a proviral or an antiviral function of ND10, we generated primary human fibroblasts with a stable, short interfering RNA-mediated knockdown (kd) of PML. In these cells, other ND10-associated proteins like hDaxx showed a diffuse nuclear distribution. Interestingly, we observed that HCMV infection induced the de novo formation of ND10-like hDaxx and Sp100 accumulations that colocalized with IE2 and were disrupted, in the apparent absence of PML, in an IE1-dependent manner during the first hours after infection. Furthermore, infection of PML-kd cells with wild-type HCMV at a low multiplicity of infection resulted in enhanced replication. In particular, a significantly increased plaque formation was detected, suggesting that more cells are able to support initiation of replication in the absence of PML. While there was no difference in viral DNA uptake between PML-kd and control cells, we observed a considerable increase in the number of immediate-early (IE) protein-positive cells, indicating that the depletion of PML augments the initiation of viral IE gene expression. These results strongly suggest that PML functions as part of an intrinsic immune mechanism against cytomegalovirus infections.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/fisiología , Regulación Viral de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Replicación Viral/genética , Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos Nucleares/análisis , Antígenos Nucleares/metabolismo , Autoantígenos/análisis , Autoantígenos/metabolismo , Núcleo Celular/química , Núcleo Celular/metabolismo , Núcleo Celular/virología , Proteínas Co-Represoras , Fibroblastos/química , Fibroblastos/metabolismo , Fibroblastos/virología , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Chaperonas Moleculares , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas Nucleares/análisis , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteína de la Leucemia Promielocítica , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
13.
J Virol ; 80(16): 7995-8005, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16873256

RESUMEN

Promyelocytic leukemia (PML) nuclear bodies (also known as ND10) are nuclear substructures that contain several proteins, including PML itself, Sp100, and hDaxx. PML has been implicated in many cellular processes, and ND10 are frequently associated with the replicating genomes of DNA viruses. During herpes simplex virus type 1 (HSV-1) infection, the viral regulatory protein ICP0 localizes to ND10 and induces the degradation of PML, thereby disrupting ND10 and dispersing their constituent proteins. ICP0-null mutant viruses are defective in PML degradation and ND10 disruption, and concomitantly they initiate productive infection very inefficiently. Although these data are consistent with a repressive role for PML and/or ND10 during HSV-1 infection, evidence in support of this hypothesis has been inconclusive. By use of short interfering RNA technology, we demonstrate that depletion of PML increases both gene expression and plaque formation by an ICP0-negative HSV-1 mutant, while having no effect on wild-type HSV-1. We conclude that PML contributes to a cellular antiviral repression mechanism that is countered by the activity of ICP0.


Asunto(s)
Herpesvirus Humano 1/genética , Proteínas Inmediatas-Precoces/metabolismo , Proteínas de Neoplasias/fisiología , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos Nucleares/análisis , Antígenos Nucleares/metabolismo , Autoantígenos/análisis , Autoantígenos/metabolismo , Proteínas Co-Represoras , Fibroblastos/química , Fibroblastos/metabolismo , Expresión Génica , Regulación Viral de la Expresión Génica , Genoma Viral , Herpesvirus Humano 1/metabolismo , Humanos , Proteínas Inmediatas-Precoces/análisis , Proteínas Inmediatas-Precoces/genética , Chaperonas Moleculares , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas Nucleares/análisis , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , ARN Interferente Pequeño/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/análisis , Ubiquitina-Proteína Ligasas/genética
14.
J Virol ; 80(11): 5423-34, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16699023

RESUMEN

We previously showed that open reading frame (ORF) UL26 of human cytomegalovirus, a member of the US22 multigene family of betaherpesviruses, encodes a novel tegument protein, which is imported into cells in the course of viral infection. Moreover, we demonstrated that pUL26 contains a strong transcriptional activation domain and is capable of stimulating the major immediate-early (IE) enhancer-promoter. Since this suggested an important function of pUL26 during the initiation of the viral replicative cycle, we sought to ascertain the relevance of pUL26 by construction of a viral deletion mutant lacking the UL26 ORF using the bacterial artificial chromosome mutagenesis procedure. The resulting deletion virus was verified by PCR, enzyme restriction, and Southern blot analyses. After infection of human foreskin fibroblasts, the UL26 deletion mutant showed a small-plaque phenotype and replicated to significantly lower titers than wild-type or revertant virus. In particular, we noticed a striking decrease of infectious titers 7 days postinfection in a multistep growth experiment, whereas the release of viral DNA from infected cells was not impaired. A further investigation of this aspect revealed a significantly diminished stability of viral particles derived from the UL26 deletion mutant. Consistent with this, we observed that the tegument composition of the deletion mutant deviates from that of the wild-type virus. We therefore hypothesize that pUL26 plays a role not only in the onset of IE gene transcription but also in the assembly of the viral tegument layer in a stable and correct manner.


Asunto(s)
Citomegalovirus/fisiología , Fosfoproteínas/química , Proteínas Virales/biosíntesis , Proteínas Virales/química , Células Cultivadas , Citomegalovirus/química , Citomegalovirus/genética , Fibroblastos/virología , Humanos , Sistemas de Lectura Abierta , Fosfoproteínas/genética , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA