Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
J Virol ; 88(4): 2337-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24307587

ABSTRACT

To facilitate studies of herpes simplex virus 1 latency, cell culture models of quiescent or latent infection have been developed. Using deep sequencing, we analyzed the expression of viral microRNAs (miRNAs) in two models employing human fibroblasts and one using rat neurons. In all cases, the expression patterns differed from that in productively infected cells, with the rat neuron pattern most closely resembling that found in latently infected human or mouse ganglia in vivo.


Subject(s)
Herpes Simplex/metabolism , Herpesvirus 1, Human/genetics , MicroRNAs/metabolism , Virus Latency/genetics , Animals , Cell Culture Techniques , Fibroblasts/metabolism , Herpes Simplex/genetics , Herpesvirus 1, Human/metabolism , High-Throughput Nucleotide Sequencing , Humans , Mice , Neurons/metabolism , Rats
3.
PLoS One ; 6(10): e25152, 2011.
Article in English | MEDLINE | ID: mdl-22003387

ABSTRACT

Alzheimer's disease (AD) afflicts around 20 million people worldwide and so there is an urgent need for effective treatment. Our research showing that herpes simplex virus type 1 (HSV1) is a risk factor for AD for the brains of people who possess a specific genetic factor and that the virus causes accumulation of key AD proteins (ß-amyloid (Aß) and abnormally phosphorylated tau (P-tau)), suggests that anti-HSV1 antiviral agents might slow AD progression. However, currently available antiviral agents target HSV1 DNA replication and so might be successful in AD only if Aß and P-tau accumulation depend on viral DNA replication. Therefore, we investigated firstly the stage(s) of the virus replication cycle required for Aß and P-tau accumulation, and secondly whether antiviral agents prevent these changes using recombinant strains of HSV1 that progress only partly through the replication cycle and antiviral agents that inhibit HSV1 DNA replication. By quantitative immunocytochemistry we demonstrated that entry, fusion and uncoating of HSV1, are insufficient to induce Aß and P-tau production. We showed also that none of the "immediate early" viral proteins is directly responsible, and that Aß and P-tau are produced at a subsequent stage of the HSV1 replication cycle. Importantly, the anti-HSV1 antiviral agents acyclovir, penciclovir and foscarnet reduced Aß and P-tau accumulation, as well as HSV1, with foscarnet being less effective in each case. P-tau accumulation was found to depend on HSV1 DNA replication, whereas Aß accumulation was not. The antiviral-induced decrease in Aß is attributable to the reduced number of new viruses, and hence the reduction in viral spread. Since antiviral agents reduce greatly Aß and P-tau accumulation in HSV1-infected cells, they would be suitable for treating AD with great advantage unlike current AD therapies, only the virus, not the host cell, would be targeted.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Antiviral Agents/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , tau Proteins/biosynthesis , Acyclovir/pharmacology , Amyloid beta-Peptides/metabolism , Animals , Cell Line , Cells, Cultured , Cricetinae , DNA Replication/drug effects , DNA, Viral/biosynthesis , Herpesvirus 1, Human/genetics , Humans , Protein Biosynthesis/drug effects , Time Factors , Virus Replication/drug effects , tau Proteins/metabolism
4.
Virology ; 414(1): 83-90, 2011 May 25.
Article in English | MEDLINE | ID: mdl-21481907

ABSTRACT

The cellular protein hypoxia-inducible factor 1 alpha (HIF-1α) was induced after infection of human fibroblasts with human cytomegalovirus (HCMV). HCMV irradiated with ultraviolet light (uv-HCMV) also elicited the effect, demonstrating that the response was provoked by interaction of the infecting virion with the cell and that viral gene expression was not required. Although induction of HIF-1α was initiated by an early event, accumulation of the protein was not detected until 9 hours post infection, with levels increasing thereafter. Infection with uv-HCMV resulted in increased abundance of HIF-1α-specific RNA, indicating stimulation of transcription. In addition, greater phosphorylation of the protein kinase Akt was observed, and the activity of this enzyme was required for induction of HIF-1α to occur. HIF-1α controls the expression of many cellular gene products; therefore the findings reveal new ways in which interaction of the HCMV particle with the host cell may cause significant alterations to cellular physiology.


Subject(s)
Cytomegalovirus/pathogenicity , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Virus Attachment , Cells, Cultured , Fibroblasts/virology , Host-Pathogen Interactions , Humans , RNA, Messenger/biosynthesis , Transcriptional Activation
5.
J Virol ; 85(9): 4212-21, 2011 May.
Article in English | MEDLINE | ID: mdl-21325419

ABSTRACT

The infection of human fetal foreskin fibroblasts (HFFF2) with human cytomegalovirus (HCMV) resulted in the induction of autophagy. This was demonstrated by the increased lipidation of microtubule-associated protein 1 light chain 3 (LC3), a hallmark of autophagy, and by the visualization of characteristic vesicles within infected cells. The response was detected first at 2 h postinfection and persisted for at least 3 days. De novo protein synthesis was not required for the effect, since HCMV that was irradiated with UV light also elicited the response, and furthermore the continuous presence of cycloheximide did not prevent induction. Infection with herpes simplex virus type 1 (HSV-1) under conditions that inhibited viral gene expression provoked autophagy, whereas UV-irradiated respiratory syncytial virus did not. The induction of autophagy occurred when cells were infected with HCMV or HSV-1 that was gradient purified, but HCMV dense bodies and HSV-1 light particles, each of which lack nucleocapsids and genomes, were inactive. The depletion of regulatory proteins Atg5 and Atg7, which are required for autophagy, reduced LC3 modification in response to infection but did not result in any detectable difference in viral or cellular gene expression at early times after infection. The electroporation of DNA into HFFF2 cultures induced the lipidation of LC3 but double-stranded RNA did not, even though both agents stimulated an innate immune response. The results show a novel, early cellular response to the presence of the incoming virion and additionally demonstrate that autophagy can be induced by the presence of foreign DNA within cells.


Subject(s)
Autophagy , Cytomegalovirus/pathogenicity , Fibroblasts/immunology , Fibroblasts/virology , Herpesvirus 1, Human/pathogenicity , Cells, Cultured , Humans , Respiratory Syncytial Viruses/pathogenicity
6.
Virus Res ; 157(1): 47-53, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21310198

ABSTRACT

The control of human cytomegalovirus (HCMV) immediate early (IE) gene expression in infected human fibroblasts was compared with that in the U2OS human osteosarcoma cells. Viral IE expression was stimulated by the virion protein pp71 and repressed by the cell protein hDaxx in fibroblasts, as expected from published data. Neither of these events occurred in infected U2OS cells, suggesting that this cell line lacks one or more factors that repress HCMV IE expression. The chromatin remodeling factor ATRX is absent from U2OS cells, therefore the effect of introducing this protein by electroporation of plasmid DNA was investigated. Provision of ATRX inhibited HCMV IE expression, and the presence of the HCMV-specified virion phosphoprotein pp71 overcame the repression. The experiments demonstrate that ATRX can act as a cellular intrinsic antiviral defense in U2OS cells by blocking gene expression from incoming HCMV genomes. In contrast, ATRX did not affect the replication of herpes simplex virus type 1, showing that there are differences in the way U2OS cells respond to the presence of the herpesviral genomes.


Subject(s)
Cytomegalovirus/genetics , DNA Helicases/metabolism , Genes, Immediate-Early , Nuclear Proteins/metabolism , Osteosarcoma/metabolism , Viral Proteins/genetics , Cell Line, Tumor , Chromatin/metabolism , Cytomegalovirus/physiology , Gene Expression , Gene Expression Regulation, Viral , Herpesvirus 1, Human/physiology , Humans , Osteosarcoma/virology , Viral Proteins/metabolism , Virion/genetics , Virion/physiology , Virus Replication , X-linked Nuclear Protein
7.
Virol J ; 6: 65, 2009 May 27.
Article in English | MEDLINE | ID: mdl-19473490

ABSTRACT

BACKGROUND: Human cytomegalovirus (HCMV) is a betaherpesvirus that causes severe disease in situations where the immune system is immature or compromised. HCMV immediate early (IE) gene expression is stimulated by the virion phosphoprotein pp71, encoded by open reading frame (ORF) UL82, and this transactivation activity is important for the efficient initiation of viral replication. It is currently recognized that pp71 acts to overcome cellular intrinsic defences that otherwise block viral IE gene expression, and that interactions of pp71 with the cell proteins Daxx and ATRX are important for this function. A further property of pp71 is the ability to enable prolonged gene expression from quiescent herpes simplex virus type 1 (HSV-1) genomes. Non-human primate cytomegaloviruses encode homologs of pp71, but there is currently no published information that addresses their effects on gene expression and modes of action. RESULTS: The UL82 homolog encoded by simian cytomegalovirus (SCMV), strain Colburn, was identified and cloned. This ORF, named S82, was cloned into an HSV-1 vector, as were those from baboon, rhesus monkey and chimpanzee cytomegaloviruses. The use of an HSV-1 vector enabled expression of the UL82 homologs in a range of cell types, and permitted investigation of their abilities to direct prolonged gene expression from quiescent genomes. The results show that all UL82 homologs activate gene expression, and that neither host cell type nor promoter target sequence has major effects on these activities. Surprisingly, the UL82 proteins specified by non-human primate cytomegaloviruses, unlike pp71, did not direct long term expression from quiescent HSV-1 genomes. In addition, significant differences were observed in the intranuclear localization of the UL82 homologs, and in their effects on Daxx. Strikingly, S82 mediated the release of Daxx from nuclear domain 10 substructures much more rapidly than pp71 or the other proteins tested. All UL82 homologs stimulated the early release of ATRX from nuclear domain 10. CONCLUSION: All of the UL82 homolog proteins analysed activated gene expression, but surprising differences in other aspects of their properties were revealed. The results provide new information on early events in infection with cytomegaloviruses.


Subject(s)
Cytomegalovirus/physiology , Gene Expression Regulation, Viral , Trans-Activators/physiology , Viral Proteins/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cell Nucleus/chemistry , Cloning, Molecular , Co-Repressor Proteins , DNA Helicases/metabolism , Genetic Vectors , Herpesvirus 1, Human/genetics , Humans , Macaca mulatta/virology , Mice , Molecular Chaperones , Nuclear Proteins/metabolism , Pan troglodytes/virology , Papio/virology , Phylogeny , Sequence Homology , X-linked Nuclear Protein
8.
PLoS Pathog ; 5(3): e1000352, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19325890

ABSTRACT

The mechanism controlling the exit from herpes simplex virus latency (HSV) is of central importance to recurrent disease and transmission of infection, yet interactions between host and viral functions that govern this process remain unclear. The cascade of HSV gene transcription is initiated by the multifunctional virion protein VP16, which is expressed late in the viral replication cycle. Currently, it is widely accepted that VP16 transactivating function is not involved in the exit from latency. Utilizing the mouse ocular model of HSV pathogenesis together with genetically engineered viral mutants and assays to quantify latency and the exit from latency at the single neuron level, we show that in vivo (i) the VP16 promoter confers distinct regulation critical for viral replication in the trigeminal ganglion (TG) during the acute phase of infection and (ii) the transactivation function of VP16 (VP16TF) is uniquely required for the exit from latency. TG neurons latently infected with the VP16TF mutant in1814 do not express detectable viral proteins following stress, whereas viruses with mutations in the other major viral transcription regulators ICP0 and ICP4 do exit the latent state. Analysis of a VP16 promoter/reporter mutant in the background of in1814 demonstrates that the VP16 promoter is activated in latently infected neurons following stress in the absence of other viral proteins. These findings support the novel hypothesis that de novo expression of VP16 regulates entry into the lytic program in neurons at all phases of the viral life cycle. HSV reactivation from latency conforms to a model in which stochastic derepression of the VP16 promoter and expression of VP16 initiates entry into the lytic cycle.


Subject(s)
Herpes Simplex Virus Protein Vmw65/genetics , Neurons/virology , Simplexvirus/physiology , Virus Latency/genetics , Animals , Gene Expression Regulation, Viral , Immunohistochemistry , Male , Mice , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transcriptional Activation , Virus Replication/genetics
9.
J Virol ; 82(24): 12543-54, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18922870

ABSTRACT

The human cytomegalovirus (HCMV) tegument protein pp71, encoded by gene UL82, stimulates viral immediate-early (IE) transcription. pp71 interacts with the cellular protein hDaxx at nuclear domain 10 (ND10) sites, resulting in the reversal of hDaxx-mediated repression of viral transcription. We demonstrate that pp71 displaces an hDaxx-binding protein, ATRX, from ND10 prior to any detectable effects on hDaxx itself and that this event contributes to the role of pp71 in alleviating repression. Introduction of pp71 into cells by transfection, infection with a pp71-expressing herpes simplex virus type 1 vector, or by generation of transformed cell lines promoted the rapid relocation of ATRX from ND10 to the nucleoplasm without alteration of hDaxx levels or localization. A pp71 mutant protein unable to interact with hDaxx did not affect the intranuclear distribution of ATRX. Infection with HCMV at a high multiplicity of infection resulted in rapid displacement of ATRX from ND10, the effect being observed maximally by 2 h after adsorption, whereas infection with the UL82-null HCMV mutant ADsubUL82 did not affect ATRX localization even at 7 h postinfection. Cell lines depleted of ATRX by transduction with shRNA-expressing lentiviruses supported increased IE gene expression and virus replication after infection with ADsubUL82, demonstrating that ATRX has a role in repressing IE transcription. The results show that ATRX, in addition to hDaxx, is a component of cellular intrinsic defenses that limit HCMV IE transcription and that displacement of ATRX from ND10 by pp71 is important for the efficient initiation of viral gene expression.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , Cytomegalovirus/metabolism , DNA Helicases/metabolism , Nuclear Proteins/metabolism , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Line , Co-Repressor Proteins , Cytomegalovirus/genetics , DNA Helicases/genetics , Gene Deletion , Gene Expression Regulation , Humans , Molecular Chaperones , Nuclear Proteins/genetics , Time Factors , Transcription, Genetic/genetics , Viral Proteins/genetics , X-linked Nuclear Protein
10.
J Virol ; 82(23): 11775-83, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18799580

ABSTRACT

Herpes simplex virus type 1 (HSV-1) mutants impaired in the activities of the structural protein VP16 and the immediate-early (IE) proteins ICP0 and ICP4 establish a quiescent infection in human fibroblasts, with most cells retaining an inactive, repressed viral genome for sustained periods in culture. To date, the quiescent state has been considered stable, since it has been reversed only by provision of herpesviral proteins, such as ICP0, not by alteration of the cell physiological state. We report that the interaction of HSV-1 with human fibroblasts can be altered significantly by transient treatment of cultures with sodium arsenite, an inducer of heat shock and oxidative stress, or gramicidin D, a toxin that selectively permeabilizes cell membranes, prior to infection. These regimens stimulated gene expression from IE-deficient HSV-1 mutants in a promoter sequence-independent manner and also overcame the replication defect of ICP0-null mutants. Reactivation of gene expression from quiescent HSV-1 genomes and the resumption of virus replication were observed following addition of arsenite or gramicidin D to cultures. Both agents induced reorganization of nuclear domain 10 structures, the sites of quiescent genomes, but appeared to do so through different mechanisms. The results demonstrate that the physiological state of the cell is important in determining the outcome of infection with IE-deficient HSV-1 and show novel methods for reactivating quiescent HSV-1 in fibroblasts with a high efficiency.


Subject(s)
Genome, Viral , Herpesvirus 1, Human/genetics , Immediate-Early Proteins/physiology , Stress, Physiological , Ubiquitin-Protein Ligases/physiology , Apoptosis , Arsenites/pharmacology , Cells, Cultured , Cytomegalovirus/genetics , Genes, Immediate-Early , Gramicidin/pharmacology , Herpesvirus 1, Human/physiology , Humans , Promoter Regions, Genetic , Virus Activation
11.
J Gen Virol ; 89(Pt 1): 68-77, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18089730

ABSTRACT

In the current study, it was shown that repressed virus genomes in quiescently infected MRC5 cells adopt a repressed histone-associated structure marked by the enrichment of deacetylated histones at a wide variety of herpes simplex virus type 1 (HSV-1) promoters. In addition, it was shown that genome de-repression, mediated by HSV-2 superinfection or delivery of ICP0 using a recombinant adenovirus vector, resulted in the enrichment of acetylated histones on HSV DNA. These data indicate that ICP0-mediated genome de-repression is intimately linked to enrichment of acetylated histones at virus promoters. The fold change in association of pan-acetylated histone H3 following Ad.TRE.ICP0-mediated de-repression consistently revealed promoter-specific variation, with the highest fold changes (>50-fold) being observed at the latency-associated transcript promoter and enhancer regions. Chromatin immunoprecipitation analyses using an antibody specific to the C terminus of histone H3 as a surrogate measure of nucleosome occupancy revealed little variability in the total loading of histone H3 at the various HSV promoters. This observation suggests that acetylation of histone H3 in response to ICP0 expression is not uniformly targeted across the HSV-1 genome during ICP0-mediated de-repression.


Subject(s)
Gene Expression Regulation, Viral , Genome, Viral , Herpesvirus 1, Human/genetics , Histones/metabolism , Immediate-Early Proteins/genetics , Ubiquitin-Protein Ligases/genetics , CD8-Positive T-Lymphocytes/virology , Cell Line , Dendritic Cells/immunology , Dendritic Cells/virology , Fibroblasts/virology , Herpesvirus 1, Human/physiology , Humans , Polymerase Chain Reaction , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/metabolism , Virus Activation
12.
J Virol ; 81(20): 10991-1004, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17670833

ABSTRACT

Herpes simplex virus type 1 (HSV-1) genomes become associated with structures related to cellular nuclear substructures known as ND10 or promyelocytic leukemia nuclear bodies during the early stages of lytic infection. This paper describes the relationship between HSV-1 genomes and ND10 in human fibroblasts that maintain the viral genomes in a quiescent state. We report that quiescent HSV-1 genomes detected by fluorescence in situ hybridization (FISH) are associated with enlarged ND10-like structures, frequently such that the FISH-defined viral foci are apparently enveloped within a sphere of PML and other ND10 proteins. The number of FISH viral foci in each quiescently infected cell is concordant with the input multiplicity of infection, with each structure containing no more than a small number of viral genomes. A proportion of the enlarged ND10-like foci in quiescently infected cells contain accumulations of the heterochromatin protein HP1 but not other common markers of heterochromatin such as histone H3 di- or trimethylated on lysine residue 9. Many of the virally induced enlarged ND10-like structures also contain concentrations of conjugated ubiquitin. Quiescent infections can be established in cells that are highly depleted for PML. However, during the initial stages of establishment of a quiescent infection in such cells, other ND10 proteins (Sp100, hDaxx, and ATRX) are recruited into virally induced foci that are likely to be associated with HSV-1 genomes. These observations illustrate that the intimate connections between HSV-1 genomes and ND10 that occur during lytic infection also extend to quiescent infections.


Subject(s)
Cell Nucleus/virology , Fibroblasts/virology , Genome, Viral , Herpesvirus 1, Human/genetics , Cell Nucleus/pathology , Cell Nucleus/ultrastructure , Fibroblasts/pathology , Herpes Simplex , Humans , In Situ Hybridization, Fluorescence , Leukemia, Promyelocytic, Acute/pathology , Nuclear Proteins/metabolism
13.
J Virol ; 81(21): 11781-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17715242

ABSTRACT

Model systems have previously been developed in which herpes simplex virus (HSV) is retained in human fibroblasts in a nonreplicating state known as quiescence. The HSV type 1 (HSV-1) immediate-early (IE) protein ICP0, an important activator of gene expression, reactivates the quiescent genome and promotes the resumption of virus replication. Previous studies reported that infection with ICP0-null HSV-1 mutants fails to reactivate quiescent HSV, even when the mutant itself undergoes productive replication, leading to the hypothesis that quiescent genomes exist in a silent configuration in which they are shielded from trans-acting factors. I reinvestigated these findings, using HSV-1 mutants with lesions in the transcription activators VP16, ICP0, and ICP4 to establish quiescent infection at high efficiency. Superinfection with ICP0-null HSV-1 mutants at a low multiplicity of infection (MOI), so that individual plaques were formed, reactivated expression from the quiescent genome, demonstrating that the requirement for ICP0 is not absolute. The previously reported failure to observe reactivation by ICP0-null mutants was shown to be a consequence of either a low initial MOI or a high superinfecting MOI. Competition between viral genomes at the level of gene expression and virus replication, especially when ICP0 was absent, was demonstrated during reactivation and also during normal infection of human fibroblasts. The results show that the multiplicity-dependent phenotype of ICP0-null mutants limits the efficiency of reactivation at low MOIs and that competition between genomes occurs at high MOIs. The conclusion that quiescent HSV genomes are extensively silenced and intrinsically insensitive to trans-acting factors must be reevaluated.


Subject(s)
Genome, Viral , Herpesvirus 1, Human/genetics , Immediate-Early Proteins/metabolism , Mutation , Ubiquitin-Protein Ligases/metabolism , DNA, Viral/metabolism , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation, Viral , Genetic Techniques , Humans , Phenotype , Plasmids/metabolism , Viral Proteins/chemistry , Virus Latency , Virus Replication
14.
J Gen Virol ; 87(Pt 5): 1113-1121, 2006 May.
Article in English | MEDLINE | ID: mdl-16603511

ABSTRACT

Human cytomegalovirus (HCMV) immediate-early (IE) transcription is stimulated by virion phosphoprotein pp71, the product of gene UL82. It has previously been shown that pp71 interacts with the cellular protein hDaxx and, in the studies presented here, the significance of this interaction was investigated for HCMV IE gene expression. In co-transfection experiments, the presence of hDaxx increased the transcriptional response of the HCMV major IE promoter (MIEP) to pp71, but it was not possible to determine whether the effect was due to an interaction between the two proteins or to stimulation of hDaxx synthesis by pp71. The use of small interfering RNA (siRNA) in long- and short-term transfection approaches reduced intracellular hDaxx levels to no more than 3 % of normal. Infection of hDaxx-depleted cells with herpes simplex virus recombinants containing the HCMV MIEP revealed significantly greater promoter activity when hDaxx levels were minimal. Similarly, reducing intracellular hDaxx amounts resulted in greater IE gene expression during infection with an HCMV mutant lacking pp71, but had no effect on IE transcription during infection with wild-type HCMV. The results suggest that hDaxx is not important as a positive-acting factor for the stimulation of HCMV IE transcription by pp71. Instead, it appears that hDaxx acts as a repressor of IE gene expression, and it is proposed here that the interaction of pp71 with hDaxx is important to relieve repression and permit efficient initiation of productive replication.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cytomegalovirus/genetics , Immediate-Early Proteins/genetics , Nuclear Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Co-Repressor Proteins , Cytomegalovirus/metabolism , Cytomegalovirus/physiology , Down-Regulation , Gene Expression Regulation, Viral , Humans , Molecular Chaperones , Nuclear Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
15.
J Virol ; 79(1): 525-35, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15596845

ABSTRACT

The human cytomegalovirus tegument protein pp71 is important for transactivation of immediate-early (IE) gene expression and for the efficient initiation of virus replication. We have analyzed the properties of pp71 by assaying its effects on gene expression from the genome of in1312, a herpes simplex virus type 1 (HSV-1) mutant devoid of functional VP16, ICP0, and ICP4. Upon infection of human fibroblasts, in1312-derived viruses are repressed and retained in a quiescent state, but the presence of pp71 prevented the quiescent state from being attained. Reporter gene cassettes cloned into the in1312 genome, in addition to the endogenous IE promoters, remained active for at least 12 days postinfection, and infected cells were viable and morphologically normal. Cells expressing pp71 remained responsive to the HSV-1 transactivating factors VP16 and ICP4 and to trichostatin A. The C-terminal 61 amino acids, but not the LACSD motif, were required for pp71 activity. In addition to preventing attainment of quiescence, pp71 was able to disrupt the quiescent state of in1312 derivatives and promote the resumption of viral gene expression after a lag of approximately 3 days. The results extend the functional analysis of pp71 and suggest a degree of similarity with the HSV-1 IE protein ICP0. The ability to provoke slow reactivation of quiescent genomes, in conjunction with cell survival, represents a novel property for a viral structural protein.


Subject(s)
Cytomegalovirus/metabolism , Gene Expression Regulation, Viral , Herpesvirus 1, Human/metabolism , Viral Proteins/metabolism , Virus Activation , Cytomegalovirus/genetics , Fibroblasts/virology , Genome, Viral , Herpes Simplex Virus Protein Vmw65/genetics , Herpes Simplex Virus Protein Vmw65/metabolism , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mutation , Recombination, Genetic , Time Factors , Ubiquitin-Protein Ligases , Viral Proteins/genetics , Virus Latency
16.
J Gen Virol ; 83(Pt 7): 1601-1612, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12075078

ABSTRACT

The human cytomegalovirus (HCMV) tegument phosphoprotein pp71 activates viral immediate early (IE) transcription and thus has a role in initiating lytic infection. Protein pp71 stimulates expression from a range of promoters in a sequence-independent manner, and in this respect behaves similarly to the herpes simplex virus type 1 (HSV-1) IE protein ICP0. The intracellular localization of pp71 was investigated after its expression from transfected plasmids or from HSV-1 mutants constructed to produce pp71 transiently. The protein colocalized with the cell promyelocytic leukaemia (PML) protein at nuclear domain 10 (ND10) structures but, unlike ICP0, pp71 did not induce disruption of ND10. The activity of pp71 in mouse sensory neurons in vivo was investigated after co-inoculation of animals with pairs of HSV-1 mutants, one expressing pp71 and the second containing the E. coli lacZ gene controlled by various promoters. In this system, pp71 stimulated beta-galactosidase expression from a range of viral IE promoters when mice were analysed at 4 days postinoculation. At later times, expression of pp71 resulted in a reduction in numbers of neurons containing beta-galactosidase, indicating cytotoxicity or promoter shutoff. The HSV-1 latency-active promoter was not responsive to pp71, demonstrating specificity in the activity of the protein. Pp71 was as active in mice lacking both copies of the PML gene (PML-/-) as in control animals, and in PML-/- fibroblasts pp71 stimulated gene expression as effectively as in other cell types. Therefore, neither the PML protein nor the normal ND10 structure is necessary for pp71 to stimulate gene expression.


Subject(s)
Cytomegalovirus/metabolism , Gene Expression Regulation, Viral , Nuclear Proteins , Viral Proteins/metabolism , Animals , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytomegalovirus/chemistry , Female , Genes, Immediate-Early , Genetic Vectors , Herpesvirus 1, Human/genetics , Humans , Lac Operon , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neurons/metabolism , Plasmids , Promyelocytic Leukemia Protein , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor Proteins , Vero Cells/metabolism , Viral Proteins/analysis , beta-Galactosidase/metabolism
17.
J Gen Virol ; 81(Pt 9): 2215-2218, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10950979

ABSTRACT

Previous studies have shown that infection of human fibroblasts with human cytomegalovirus (HCMV) results in activation of cellular interferon-responsive gene expression. We demonstrate here that infection of human fibroblasts with herpes simplex virus type 1 (HSV-1) in the absence of de novo protein synthesis also induces the expression of interferon-responsive genes. Five genes tested (encoding ISG54, IFI56, ISG15, 9-27 and MxA) were activated by infection with HSV-1, although the degree of response varied between the individual genes. HSV-1 was a less efficient inducer than HCMV. The effect was a consequence of binding of the virus particle to the cell surface or of the presence of virion components within the infected cell. Induction was mediated by a pathway other than the mechanism through which interferon-alpha mediates its effects on cellular gene expression.


Subject(s)
Carrier Proteins , GTP-Binding Proteins , Gene Expression Regulation, Viral , Herpes Simplex/genetics , Herpesvirus 1, Human , Interferons/physiology , Ubiquitins/analogs & derivatives , Adaptor Proteins, Signal Transducing , Antiviral Agents/biosynthesis , Cell Line , Cytokines/biosynthesis , Fibroblasts/virology , Humans , Leucine Zippers , Myxovirus Resistance Proteins , Protein Biosynthesis , RNA-Binding Proteins , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...