Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Virol ; 85(23): 12631-7, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21937654

RESUMEN

In cell culture experiments, phosphorylation appears to be a critical regulator of the herpes simplex virus 1 (HSV-1) immediate-early (IE) protein, ICP0, which is an E3 ubiquitin ligase that transactivates viral gene expression. Three major regions of phosphorylation in ICP0 (amino acids 224 to 232, 365 to 371, and 508 to 518) have been identified, and mutant viruses that block phosphorylation sites within each region (termed Phos 1, 2, and 3, respectively) have been constructed. Previous studies indicated that replication of Phos 1 is significantly reduced compared to that of wild-type virus in cell culture (C. Boutell, et al., J. Virol. 82:10647-10656, 2008). To determine the effects these phosphorylation site mutations have on the viral life cycle in vivo, mice were ocularly infected with wild-type HSV-1, the Phos mutants, or their marker rescue counterparts. Subsequently, viral replication, establishment of latency, and viral explant-induced reactivation of these viruses were examined. Relative to wild-type virus, Phos 1 eye titers were reduced as much as 7- and 18-fold on days 1 and 5 postinfection, respectively. Phos 2 eye titers showed a decrease of 6-fold on day 1 postinfection. Titers of Phos 1 and 2 trigeminal ganglia were reduced as much as 16- and 20-fold, respectively, on day 5 postinfection. Additionally, the reactivation efficiencies of Phos 1 and 2 were impaired relative to wild-type HSV-1, although both viruses established wild-type levels of latency in vivo. The acute replication, latency, and reactivation phenotypes of Phos 3 were similar to those of wild-type HSV-1. We conclude from these studies that phosphorylation is likely a key modulator of ICP0's biological activities in a mouse ocular model of HSV-1 infection.


Asunto(s)
Oftalmopatías/virología , Herpes Simple/virología , Herpesvirus Humano 1/patogenicidad , Proteínas Inmediatas-Precoces/genética , Mutación/genética , Ganglio del Trigémino/virología , Ubiquitina-Proteína Ligasas/genética , Activación Viral , Replicación Viral , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Oftalmopatías/metabolismo , Femenino , Genoma Viral , Herpes Simple/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Ratones , Datos de Secuencia Molecular , Fosforilación , Homología de Secuencia de Aminoácido , Transcripción Genética , Ganglio del Trigémino/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células Vero , Latencia del Virus
2.
J Virol ; 84(1): 188-200, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19828605

RESUMEN

Herpesviruses are characterized by the ability to establish lifelong latent infections and to reactivate periodically, leading to recurrent disease. The herpes simplex virus type 1 (HSV-1) genome is maintained in a quiescent state in sensory neurons during latency, which is characterized by the absence of detectable viral protein synthesis. Cellular factors induced by stress may act directly on promoters within the latent viral genome to induce the transcription of viral genes and trigger reactivation. In order to identify which viral promoters are induced by stress and elucidate the cellular mechanism responsible for the induction, we generated a panel of HSV-1 promoter-luciferase constructs and measured their response to heat shock. Of the promoters tested, those of ICP0 and ICP22 were the most strongly upregulated after heat shock. Microarray analysis of lytically infected cells supported the upregulation of ICP0 and ICP22 promoters by heat shock. Mutagenic analysis of the ICP0 promoter identified two regions necessary for efficient heat-induced promoter activity, both containing predicted nuclear factor Y (NF-Y) sites, at bases -708 and -75 upstream of the transcriptional start site. While gel shift analysis confirmed NF-Y binding to both sites, only the site at -708 was important for efficient heat-induced activity. Reverse transcription-PCR analysis of selected viral transcripts in the presence of dominant-negative NF-Y confirmed the requirement for NF-Y in the induction of the ICP0 but not the ICP22 promoter by heat shock in lytically infected cells. These findings suggest that the immediate-early ICP0 gene may be among the first genes to be induced during the early events in HSV-1 reactivation, that NF-Y is important for this induction, and that other factors induce the ICP22 promoter.


Asunto(s)
Factor de Unión a CCAAT/fisiología , Regulación Viral de la Expresión Génica , Respuesta al Choque Térmico/genética , Herpesvirus Humano 1/genética , Proteínas Inmediatas-Precoces/genética , Regiones Promotoras Genéticas , Ubiquitina-Proteína Ligasas/genética , Animales , Línea Celular , Chlorocebus aethiops , Perfilación de la Expresión Génica , Genes Inmediatos-Precoces , Genes Virales , Humanos , Células Vero , Activación Viral
3.
J Virol ; 83(18): 9183-94, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19570862

RESUMEN

ICP22, an immediate-early protein of herpes simplex virus type 1 (HSV-1), is required for viral replication in nonpermissive cell types and for expression of a class of late viral proteins which includes glycoprotein C. An understanding of the mechanism of ICP22 function has been complicated by the coexpression of the full-length protein with an in-frame, C-terminus-specific protein, U(S)1.5. In this report, we confirm that the U(S)1.5 protein is a bona fide translation product since it is detected during infections with three laboratory strains and two low-passage clinical isolates of HSV-1. To clarify the expression patterns of the ICP22 and U(S)1.5 proteins, we examined their synthesis from plasmids in transient expression assays. Because previous studies had identified two different U(S)1.5 translational start sites, we attempted to determine which is correct by studying the effects of a series of deletion, nonsense, and methionine substitutions on U(S)1.5 expression. First, amino acids 90 to 420 encoded by the ICP22 open reading frame (ORF) migrated at the mobility of U(S)1.5 in sodium dodecyl sulfate-polyacrylamide gels. Second, introduction of a stop codon downstream of M90 ablated expression of both ICP22 and U(S)1.5. Finally, mutation of M90 to alanine (M90A) allowed expression of full-length ICP22 while dramatically reducing expression of U(S)1.5. Levels of U(S)1.5 but not ICP22 protein expression were also reduced in cells infected with an M90A mutant virus. Thus, we conclude that expression of IC22 and that of U(S)1.5 can occur independently of each other and that U(S)1.5 translation initiates at M90 of the ICP22 ORF.


Asunto(s)
Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/genética , Biosíntesis de Proteínas , Proteínas Virales/biosíntesis , Mutación , Sistemas de Lectura Abierta/genética , Proteínas Reguladoras y Accesorias Virales/genética
4.
J Virol ; 83(17): 8733-43, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19535441

RESUMEN

Of the five herpes simplex virus type 1 immediate early (IE) proteins, the least is known about the function of ICP22 during productive infection and latency. Research characterizing the physical and functional properties of the protein has been limited because ICP22 has proven to be difficult to express in transient assays. In addition, genetic analysis of ICP22 has been complicated by the fact that the C terminus of ICP22 is expressed as a discrete protein product. In order to characterize properties of mutant and wild-type ICP22, we developed a transient expression system. We found that ICP22 can be expressed at detectable levels when placed under the control of the cytomegalovirus IE promoter, confirming recent observations by K. A. Fraser and S. A. Rice (J. Virol. 81:5091-5101, 2007). We extended this analysis to show that ICP22 can also be expressed from its own promoter in the presence of other viral factors, either by coexpression with ICP0 or by infection with an ICP22 null virus. Notably, infection of cells transfected with an ICP22 expression vector yielded ICP22 protein that was modified in a manner similar to that of ICP22 protein detected in wild-type-infected cells. We go on to demonstrate that the failure of ICP22 protein to be expressed in transiently transfected cells was not due to inactivity of the ICP22 promoter, but rather to the ability of ICP22 to inhibit expression of reporter gene activity, including its own, in transient assays. Of special note was the observation that expression of ICP22 was sufficient to prevent transactivation of reporter genes by ICP0. Finally, transient expression of ICP22 was sufficient to complement replication of an ICP22 null virus, demonstrating that this system can be used to study functional properties of ICP22. Collectively, this transient expression system facilitates tests of the physical and functional properties of ICP22 and ICP22 mutants prior to introduction of mutant genes into the viral genome.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/fisiología , Replicación Viral , Eliminación de Gen , Prueba de Complementación Genética , Herpesvirus Humano 1/genética , Proteínas Inmediatas-Precoces/genética , Regiones Promotoras Genéticas , Ubiquitina-Proteína Ligasas/fisiología
5.
J Virol ; 82(22): 11472-5, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18768961

RESUMEN

Isoleucine deprivation of cellular monolayers prior to infection has been reported to result in partial complementation of a herpes simplex virus type 1 (HSV-1) ICP0 null (ICP0(-)) mutant. We now report that glutamine deprivation alone is able to enhance the plating efficiency of an ICP0(-) virus and that isoleucine deprivation has little or no effect. Because a low glutamine level is associated with stress and because stress is known to induce reactivation, low levels of glutamine may be relevant to the reactivation of HSV-1 from latency. Additionally, we demonstrate that arginine and methionine deprivation result in partial complementation of the ICP0(-) virus.


Asunto(s)
Glutamina/metabolismo , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 1/genética , Proteínas Inmediatas-Precoces/genética , Mutación , Ubiquitina-Proteína Ligasas/genética , Animales , Chlorocebus aethiops , Células Vero , Ensayo de Placa Viral
6.
Nat Rev Drug Discov ; 2(4): 278-88, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12669027

RESUMEN

In the absence of effective vaccines to control herpesvirus infections, nucleosidic antiviral drugs have been the mainstay of clinical treatment since their development in the late 1970s. However, given the drawbacks of these drugs, including the increasing emergence of drug-resistant clinical isolates, new strategies for treating herpesvirus infections are warranted. A range of promising new drugs with novel molecular targets has been developed, but will they cure latent infections?


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Herpesviridae/tratamiento farmacológico , Herpesviridae/efectos de los fármacos , Farmacorresistencia Microbiana , Herpesviridae/patogenicidad , Humanos , Inhibidores de la Síntesis del Ácido Nucleico
8.
Sci Rep ; 4: 3664, 2014 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-24441171

RESUMEN

Experimental and bioinformatic studies of transcription initiation by RNA polymerase II (RNAP2) have revealed a mechanism of RNAP2 transcription initiation less uniform across gene promoters than initially thought. However, the general transcription factor TFIIB is presumed to be universally required for RNAP2 transcription initiation. Based on bioinformatic analysis of data and effects of TFIIB knockdown in primary and transformed cell lines on cellular functionality and global gene expression, we report that TFIIB is dispensable for transcription of many human promoters, but is essential for herpes simplex virus-1 (HSV-1) gene transcription and replication. We report a novel cell cycle TFIIB regulation and localization of the acetylated TFIIB variant on the transcriptionally silent mitotic chromatids. Taken together, these results establish a new paradigm for TFIIB functionality in human gene expression, which when downregulated has potent anti-viral effects.


Asunto(s)
Factor de Transcripción TFIIB/metabolismo , Acetilación , Animales , Sitios de Unión , Ciclo Celular/genética , Línea Celular , Conjuntos de Datos como Asunto , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Regulación Viral de la Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Genes Letales , Genoma Humano , Herpesvirus Humano 1/genética , Humanos , Especificidad de Órganos/genética , Unión Proteica , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , Factor de Transcripción TFIIB/deficiencia , Factor de Transcripción TFIIB/genética , Sitio de Iniciación de la Transcripción , Transcripción Genética , Transcriptoma
9.
J Virol ; 81(19): 10699-711, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17634223

RESUMEN

Two in-frame, C-terminal isoforms of the herpes simplex virus type 1 (HSV-1) origin binding protein (OBP), OBPC-1 and OBPC-2, and a unique C-terminal transcript, UL8.5, are specified by HSV-1 DNA. As the first isoform identified, OBPC-1 was initially assumed to be the product of the UL8.5 transcript. Recent evidence has demonstrated, however, that OBPC-1 is a cathepsin B-mediated cleavage product of OBP, suggesting that OBPC-2 is the product of the UL8.5 transcript. Because both OBPC-1 and -2 contain the majority of the OBP DNA binding domain, we hypothesized that both may be involved in regulating origin-dependent, OBP-mediated viral DNA replication. In this paper, we demonstrate that OBPC-2 is, indeed, the product of the UL8.5 transcript. The translational start site of OBPC-2 was mapped, and a virus (M571A) that does not express this protein efficiently was constructed. Using M571A, we have shown that OBPC-2 is able to bind origin DNA, even though it lacks seven N-terminal amino acid residues of the previously mapped OBP DNA binding domain, resulting in a revision of the limits of the OBP DNA binding domain. Consistent with their proposed roles in regulating viral DNA replication, OBPC-1 and -2 act together to down-regulate viral DNA replication in vitro. During functional studies in vivo, OBPC-2 was identified as a factor that increases mortality in the mouse ocular model of HSV-1 infection.


Asunto(s)
Replicación del ADN , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Complejo de Reconocimiento del Origen/metabolismo , Replicación Viral/genética , Animales , ADN Viral/biosíntesis , Herpesvirus Humano 1/genética , Ratones , Mutación , Complejo de Reconocimiento del Origen/genética , Biosíntesis de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Origen de Réplica , Transcripción Genética , Latencia del Virus/genética
10.
Virology ; 361(2): 372-83, 2007 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-17207829

RESUMEN

A recombinant HSV-1 virus expressing EGFP from the HCMV major immediate early promoter (KOS-CMVGFP) was constructed to monitor viral replication and spread in vitro and in mice. KOS-CMVGFP replicated as efficiently as wild-type virus, strain KOS, in single cycle growth experiments in Vero cells indicating that the recombinant virus has no significant growth defects in vitro. Following ocular inoculation of mice, KOS-CMVGFP exhibited slight but statistically significant reductions in mouse tear film titers relative to wild-type virus. Progression of virus infection of the eyes, periocular tissue, and snout was readily followed by fluorescence microscopy. Insertion of the EGFP expression cassette into the KOS genome had no effect on the efficiency of establishment of latency as determined by quantitative competitive PCR of viral genomes in latently infected TG. KOS-CMVGFP reactivated with wild-type kinetics and efficiency by explant cocultivation, but exhibited a significant delay in the kinetics and a modest reduction in the efficiency of reactivation compared to KOS in the more sensitive TG cell culture model. Notably, EGFP expression preceded the detection of infectious virus by greater than 24 h in both ex vivo models and thus is a useful marker of the early stages in the induction of reactivation.


Asunto(s)
Modelos Animales de Enfermedad , Herpes Simple/virología , Herpesvirus Humano 1 , Ratones , Animales , Células Cultivadas , Chlorocebus aethiops , Córnea/metabolismo , Córnea/virología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Herpes Simple/metabolismo , Herpes Simple/patología , Herpesvirus Humano 1/patogenicidad , Herpesvirus Humano 1/fisiología , Humanos , Masculino , Ratones Endogámicos ICR , Regiones Promotoras Genéticas , Proteínas Recombinantes/biosíntesis , Recombinación Genética , Células Vero , Virulencia , Activación Viral , Replicación Viral
11.
J Virol ; 81(17): 9175-82, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17553869

RESUMEN

Although the seven viral proteins required for herpes simplex virus type 1 (HSV-1) DNA replication have been identified, the mechanism by which viral DNA synthesis is regulated is unclear. HSV-1 DNA replication is thought to occur in two stages: origin-dependent DNA replication (stage I) mediated by the origin binding protein (OBP), followed by origin- and OBP-independent DNA replication (stage II). The mechanism that facilitates the switch from stage I to stage II is unknown; however, it must involve the loss of OBP function or OBP itself from the replication initiation complex. Previous studies from this laboratory identified a transcript (UL8.5) and protein (OBPC) that are in frame with and comprise the C terminus of the gene specifying OBP. Because of its DNA binding ability, OBPC has been hypothesized to mediate the switch from stage I to stage II. Here, we identify a second protein (OBPC-2) that is also in frame with the C terminus of OBP but comprises a smaller portion of the protein. We demonstrate that the protein originally identified (OBPC-1) is a cathepsin B-mediated cleavage product of OBP, while OBPC-2 may be the product of the UL8.5 transcript. We further demonstrate that the cleavage of OBP to yield OBPC-1 is dependent upon viral DNA replication. These results suggest that cleavage may be a mechanism by which OBP levels and/or activity are regulated during infection.


Asunto(s)
Catepsina B/metabolismo , Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 1/fisiología , Proteínas Virales/metabolismo , Replicación Viral/fisiología , Animales , Chlorocebus aethiops , Modelos Biológicos , Isoformas de Proteínas , Procesamiento Proteico-Postraduccional , Células Vero , Proteínas Virales/genética
12.
Virology ; 360(2): 305-21, 2007 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-17126870

RESUMEN

The host innate response to viral infection includes the production of interferons, which is dependent on the coordinated activity of multiple transcription factors. Herpes simplex virus 1 (HSV-1) has been shown to block efficient interferon expression by multiple mechanisms. We and others have demonstrated that HSV-1 can inhibit the transcription of genes promoted by interferon regulatory factor-3 (IRF-3), including interferon beta (IFN-beta), and that the immediate-early ICP0 protein is sufficient for this function. However, the exact mechanism by which ICP0 blocks IRF-3 activity has yet to be determined. Unlike some other viral proteins that inhibit IRF-3 activity, ICP0 does not appear to affect phosphorylation and dimerization of IRF-3. Here, we show that a portion of activated IRF-3 co-localizes with nuclear foci containing ICP0 at early times after virus infection. Co-localization to ICP0-containing foci is also seen with the IRF-3-binding partners and transcriptional co-activators, CBP and p300. In addition, using immunoprecipitation of infected cell lysates, we can immunoprecipitate a complex containing ICP0, IRF-3, and CBP. Thus we hypothesize that ICP0 recruits activated IRF-3 and CBP/p300 to nuclear structures, away from the host chromatin. This leads to the inactivation and accelerated degradation of IRF-3, resulting in reduced transcription of IFN-beta and an inhibition of the host response. Therefore, ICP0 provides an example of how viruses can block IFN-beta induction by sequestration of important transcription factors essential for the host response.


Asunto(s)
Núcleo Celular/metabolismo , Herpesvirus Humano 1/inmunología , Proteínas Inmediatas-Precoces/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/biosíntesis , Ubiquitina-Proteína Ligasas/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Western Blotting , Línea Celular , Inhibidores de Cisteína Proteinasa/farmacología , Herpesvirus Humano 1/fisiología , Humanos , Proteínas Inmediatas-Precoces/química , Inmunoprecipitación , Leupeptinas/farmacología , Microscopía Fluorescente , Ubiquitina-Proteína Ligasas/química
13.
J Virol ; 80(1): 440-50, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16352568

RESUMEN

In vitro studies of herpes simplex virus type 1 (HSV-1) viruses containing mutations in core sequences of the viral origins of DNA replication, oriL and oriS, that eliminate the ability of these origins to initiate viral-DNA synthesis have demonstrated little or no effect on viral replication in cultured cells, leading to the conclusion that the two types of origins are functionally redundant. It remains unclear, therefore, why origins that appear to be redundant are maintained evolutionarily in HSV-1 and other neurotropic alphaherpesviruses. To test the hypothesis that oriL and oriS have distinct functions in the HSV-1 life cycle in vivo, we determined the in vivo phenotypes of two mutant viruses, DoriL-I(LR) and DoriS-I, containing point mutations in oriL and oriS site I, respectively, that eliminate origin DNA initiation function. Following corneal inoculation of mice, tear film titers of DoriS-I were reduced relative to wild-type virus. In all other tests, however, DoriS-I behaved like wild-type virus. In contrast, titers of DoriL-I(LR) in tear film, trigeminal ganglia (TG), and hindbrain were reduced and mice infected with DoriL-I(LR) exhibited greatly reduced mortality relative to wild-type virus. In the TG explant and TG cell culture models of reactivation, DoriL-I(LR) reactivated with delayed kinetics and, in the latter model, with reduced efficiency relative to wild-type virus. Rescuant viruses DoriL-I(LR)-R and DoriS-I-R behaved like wild-type virus in all tests. These findings demonstrate that functional differences exist between oriL and oriS and reveal a prominent role for oriL in HSV-1 pathogenesis.


Asunto(s)
Herpesvirus Humano 1/patogenicidad , Origen de Réplica/genética , Latencia del Virus/genética , Animales , Chlorocebus aethiops , Replicación del ADN/fisiología , ADN Viral/biosíntesis , ADN Viral/genética , Herpes Simple/virología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/fisiología , Humanos , Ratones , Ratones Endogámicos ICR , Mutación Puntual , Origen de Réplica/fisiología , Células Vero , Activación Viral , Latencia del Virus/fisiología
14.
J Virol ; 80(9): 4528-37, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16611913

RESUMEN

This lab reported previously that the plating efficiency of a herpes simplex virus type 1 ICP0-null mutant was enhanced upon release from an isoleucine block which synchronizes cells to G1 phase (W. Cai and P. A. Schaffer, J. Virol. 65:4078-4090, 1991). Peak plating efficiency occurred as cells cycled out of G1 and into S phase, suggesting that the enhanced plating efficiency was due to cellular activities present in late G1/early S phase. We have found, however, that the enhanced plating efficiency did not occur when cells were synchronized by alternative methods. We now report that the plating efficiency of ICP0- viruses is not enhanced at a particular stage of the cell cycle but rather is enhanced by specific cellular stresses. Both the plating and replication efficiencies of ICP0- viruses were enhanced as much as 25-fold to levels similar to that of wild-type virus when monolayers were heat shocked prior to infection. In addition to heat shock, UV-C irradiation but not cold shock of monolayers prior to infection resulted in enhanced plating efficiency. We further report that the effect of cellular stress is transient and that cell density rather than age of the monolayers is the primary determinant of ICP0- virus plating efficiency. As both cell stress and ICP0 are required for efficient reactivation from latency, the identification of cellular activities that complement ICP0- viruses may lead to the identification of cellular activities that are important for reactivation from neuronal latency.


Asunto(s)
Ciclo Celular/fisiología , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Temperatura , Ubiquitina-Proteína Ligasas/metabolismo , Rayos Ultravioleta , Replicación Viral , Animales , Chlorocebus aethiops , Frío , Respuesta al Choque Térmico , Herpesvirus Humano 1/efectos de la radiación , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Células Vero , Replicación Viral/efectos de la radiación
15.
J Virol ; 80(8): 4005-16, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16571817

RESUMEN

Herpes simplex virus type 1 ICP22-/U(S)1.5- mutants initiate viral gene expression in all cells; however, in most cell types, the replication process stalls due to an inability to express gamma2 late proteins. Although the function of ICP22/U(S)1.5 has not been established, it has been suggested that these proteins activate, induce, or repress the activity of cellular proteins during infection. In this study, we hypothesized that cell cycle-associated proteins are targets of ICP22/U(S)1.5. For this purpose, we first isolated and characterized an ICP22-/U(S)1.5- mutant virus, 22/n199. Like other ICP22-/U(S)1.5- mutants, 22/n199 replicates in a cell-type-specific manner and fails to induce efficient gamma2 late gene expression in restrictive cells. Although synchronization of restrictive human embryonic lung cells in each phase of the cell cycle did not overcome the growth restrictions of 22/n199, synchronization of permissive Vero cells in S phase rendered them less able to support 22/n199 plaque formation and replication. Consistent with this finding, expression of cellular S-phase cyclins was altered in an ICP22/U(S)1.5-dependent manner specifically when S-phase Vero cells were infected. Collectively, these observations support the notion that ICP22/U(S)1.5 deregulates the cell cycle upon infection of S-phase permissive cells by altering expression of key cell cycle regulatory proteins either directly or indirectly.


Asunto(s)
Ciclinas/análisis , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/fisiología , Fase S , Replicación Viral , Animales , Chlorocebus aethiops , Ciclina E/fisiología , Quinasas Ciclina-Dependientes/análisis , Regulación hacia Abajo , Humanos , Células Vero , Proteínas Reguladoras y Accesorias Virales
16.
J Virol ; 80(19): 9381-90, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16973544

RESUMEN

The immediate-early regulatory protein ICP22 is required for efficient replication of herpes simplex virus type 1 in some cell types (permissive) but not in others (restrictive). In mice infected via the ocular route, the pathogenesis of an ICP22- virus, 22/n199, was altered relative to that of wild-type virus. Specifically, tear film titers of 22/n199-infected mice were significantly reduced at 3 h postinfection relative to those of mice infected with wild-type virus. Further, 22/n199 virus titers were below the level of detection in trigeminal ganglia (TG) during the first 9 days postinfection. On day 30 postinfection, TG from 22/n199-infected mice contained reduced viral genome loads and exhibited reduced expression of latency-associated transcripts and reduced reactivation efficiency relative to TG from wild-type virus-infected mice. Notably, the first detectable alteration in the pathogenesis of 22/n199 in these tests occurred in the eye prior to the onset of nascent virus production. Thus, ICP22- virions appeared to be degraded, cleared, or adsorbed more rapidly than wild-type virions, implying potential differences in the composition of the two virion types. Analysis of the protein composition of purified extracellular virions indicated that ICP22 is not a virion component and that 22/n199 virions sediment at a reduced density relative to wild-type virions. Although similar to wild-type virions morphologically, 22/n199 virions contain reduced amounts of two gamma2 late proteins, US11 and gC, and increased amounts of two immediate-early proteins, ICP0 and ICP4, as well as protein species not detected in wild-type virions. Although ICP22- viruses replicate to near-wild-type levels in permissive cells, the virions produced in these cells are biochemically and physically different from wild-type virions. These virion-specific differences in ICP22- viruses add a new level of complexity to the functional analysis of this immediate-early viral regulatory protein.


Asunto(s)
Herpes Simple/virología , Herpesvirus Humano 1/química , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Virión/química , Virión/fisiología , Animales , Línea Celular , Chlorocebus aethiops , Genoma Viral/genética , Herpes Simple/genética , Herpes Simple/patología , Humanos , Proteínas Inmediatas-Precoces/aislamiento & purificación , Cinética , Masculino , Ratones , Mutación/genética , Ganglio del Trigémino/metabolismo , Ganglio del Trigémino/patología , Ganglio del Trigémino/virología , Proteínas Reguladoras y Accesorias Virales , Virión/genética , Virión/aislamiento & purificación
17.
J Virol ; 79(20): 12783-97, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16188981

RESUMEN

Technical challenges associated with mutagenesis of the large oriL palindrome have hindered comparisons of the functional roles of the herpes simplex virus type 1 (HSV-1) origins of DNA replication, oriL and oriS, in viral replication and pathogenesis. To address this problem, we have developed a novel PCR-based strategy to introduce site-specific mutations into oriL and other large palindromes. Using this strategy, we generated three plasmids containing mutant forms of oriL, i.e., pDoriL-I(L), pDoriL-I(R), and pDoriL-I(LR), containing point mutations in the left, right, and both copies, respectively, of the origin binding protein (OBP) binding site (site I) which eliminate OBP binding. In in vitro DNA replication assays, plasmids with mutations in only one arm of the palindrome supported origin-dependent DNA replication, whereas plasmids with symmetrical mutations in both arms of the palindrome were replication incompetent. An analysis of the cloned mutant plasmids used in replication assays revealed that a fraction of each plasmid mutated in only one arm of the palindrome had lost the site I mutation. In contrast, plasmids containing symmetrical mutations in both copies of site I retained both mutations. These observations demonstrate that the single site I mutations in pDoriL-I(L) and pDoriL-I(R) are unstable upon propagation in bacteria and suggest that functional forms of both the left and right copies of site I are required to initiate DNA replication at oriL. To examine the role of oriL and oriS site I in virus replication, we introduced the two site I mutations in pDoriL-I(LR) into HSV-1 DNA to yield the mutant virus DoriL-I(LR) and the same point mutations into the single site I sequence present in both copies of oriS to yield the mutant virus DoriS-I. In Vero cells and primary rat embryonic cortical neurons (PRN) infected with either mutant virus, viral DNA synthesis and viral replication were efficient, confirming that the two origins can substitute functionally for one another in vitro. Measurement of the levels of oriL and oriS flanking gene transcripts revealed a modest alteration in the kinetics of ICP8 transcript accumulation in DoriL-I(LR)-infected PRN, but not in Vero cells, implicating a cell-type-specific role for oriL in regulating ICP8 transcription.


Asunto(s)
ADN Viral/genética , Herpesvirus Humano 1/genética , Origen de Réplica/fisiología , Animales , Secuencia de Bases , Células Cultivadas , ADN Viral/biosíntesis , Herpes Simple/virología , Herpesvirus Humano 1/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación Puntual , Ratas , Origen de Réplica/genética
18.
J Virol ; 79(2): 1232-43, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15613350

RESUMEN

The herpes simplex virus type 1 (HSV-1) immediate-early (IE) regulatory protein infected-cell protein 0 (ICP0) is a strong and global transactivator of both viral and cellular genes. In a previous study, we reported that ICP0 is highly phosphorylated and contains at least seven distinct phosphorylation signals as determined by phosphotryptic peptide mapping (D. J. Davido et al., J. Virol. 76:1077-1088, 2002). Since phosphorylation affects the activities of many viral regulatory proteins, we sought to determine whether the phosphorylation of ICP0 affects its functions. To address this question, it was first necessary to identify the regions of ICP0 that are phosphorylated. For this purpose, ICP0 was partially purified, and phosphorylation sites were mapped by microcapillary high-pressure liquid chromatography tandem mass spectrometry. Three phosphorylated regions containing 11 putative phosphorylation sites, all within or adjacent to domains important for the transactivating activity of ICP0, were identified. The 11 sites were mutated to alanine as clusters in each of the three regions by site-directed mutagenesis, generating plasmids expressing mutant forms of ICP0: Phos 1 (four mutated sites), Phos 2 (three mutated sites), and Phos 3 (four mutated sites). One-dimensional phosphotryptic peptide analysis confirmed that the phosphorylation state of each Phos mutant form of ICP0 is altered relative to that of wild-type ICP0. In functional assays, the ICP0 phosphorylation site mutations affected the subcellular and subnuclear localization of ICP0, its ability to alter the staining pattern of the nuclear domain 10 (ND10)-associated protein PML, and/or its transactivating activity in Vero cells. Only mutations in Phos 1, however, impaired the ability of ICP0 to complement the replication of an ICP0 null mutant in Vero cells. This study thus suggests that phosphorylation is an important regulator of ICP0 function.


Asunto(s)
Proteínas Inmediatas-Precoces/fisiología , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente , Proteínas Inmediatas-Precoces/análisis , Espectrometría de Masas , Datos de Secuencia Molecular , Mutación , Fosforilación , Activación Transcripcional , Ubiquitina-Proteína Ligasas , Células Vero
19.
J Virol ; 76(3): 1077-88, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11773384

RESUMEN

The cyclin-dependent kinase (cdk) inhibitor Roscovitine (Rosco) reduces transcription of herpes simplex virus early genes significantly, even in the presence of wild-type levels of immediate-early (IE) viral proteins, suggesting that the transactivating functions of IE proteins may require the activities of one or more Rosco-sensitive cdk (L. M. Schang, A. Rosenberg, and P. A. Schaffer, J. Virol. 73:2161-2172, 1999). Based on this observation, we sought to determine whether Rosco alters the transactivating activity and posttranslational modification state of the IE protein, infected cell protein 0 (ICP0), in KOS6beta-infected Vero cells. KOS6beta is a KOS-derived recombinant virus containing an ICP0-inducible ICP6 promoter::lacZ cassette. To monitor ICP0's transactivating activity, KOS6beta-infected cells were released from a cycloheximide (CHX)-mediated protein synthesis block into medium with or without Rosco, and beta-galactosidase activity was measured. Rosco inhibited the ability of ICP0 to transactivate the ICP6 promoter by 50-fold. This inhibition was shown not to be a consequence of inhibition of ICP6 basal promoter activity or aberrant nuclear localization of ICP0. Rosco also altered the electrophoretic mobility of a portion of ICP0 molecules derived from KOS-infected cells following reversal of a CHX block. Notably, however, Rosco had only a minimal effect on the phosphorylation state of ICP0. We conclude that ICP0's transactivating activity requires Rosco-sensitive cdks and hypothesize that these cdks regulate the functions of cellular enzymes which modify ICP0, and are, consequently, required for its transactivating activity. Thus, we propose that Rosco regulates ICP0's posttranslational state by mechanisms other than, or in addition to, phosphorylation.


Asunto(s)
Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Proteínas Inmediatas-Precoces/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Purinas/farmacología , Transactivadores/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Animales , Núcleo Celular/metabolismo , Chlorocebus aethiops , Electroforesis en Gel de Poliacrilamida , Expresión Génica , Ingeniería Genética , Herpesvirus Humano 1/genética , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Mapeo Peptídico , Fosfoproteínas/análisis , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , Roscovitina , Transactivadores/metabolismo , Tripsina , Ubiquitina-Proteína Ligasas , Células Vero , Proteínas Virales/genética , beta-Galactosidasa/genética
20.
J Virol ; 76(15): 7724-35, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12097586

RESUMEN

Herpes simplex virus (HSV) establishes productive (lytic) infections in nonneuronal cells and nonproductive (latent) infections in neurons. It has been proposed that HSV establishes latency because quiescent neurons lack cellular factors required for productive infection. It has been further proposed that these putative factors are induced following neuronal stress, as a requirement for HSV reactivation. To date, the identity of these putative cellular factors remains unknown. We have demonstrated that cyclin-dependent kinase (cdk) 1, 2, or 7 is required for HSV replication in nonneuronal cells. Interestingly, cdks 1 and 2 are not expressed in quiescent neurons but can be induced in stressed neurons. Thus, cdks may be among the cellular proteins required for HSV reactivation whose neuronal expression is differentially regulated during stress. Herein, we determined that neuronal expression of nuclear cdk2, cdk4, and cyclins E and D2 (which activate cdks 2 and 4, respectively) was induced following explant cultivation, a stressful stimulus that induces HSV reactivation. In contrast, neuronal expression of cdk7 and cytoplasmic cdk4 decreased during explant cultivation, whereas cdk3 was detected in the same small percentage of neurons before and after explant cultivation and cdks 1, 5, and 6 were not detected in neuronal cell bodies. HSV-1 reactivated specifically in neurons expressing nuclear cdk2 and cdk4, and an inhibitor specific for cdk2 inhibited HSV-1 reactivation. We conclude that neuronal levels of cdk2 are among the factors that determine the outcome of HSV infections of neurons.


Asunto(s)
Quinasas CDC2-CDC28 , Quinasas Ciclina-Dependientes/metabolismo , Neuronas/virología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas , Simplexvirus/crecimiento & desarrollo , Activación Viral , Animales , Núcleo Celular/metabolismo , Técnicas de Cultivo/métodos , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Ciclinas/metabolismo , Inmunohistoquímica , Queratitis Herpética/virología , Ratones , Ratones Endogámicos ICR , Ganglio del Trigémino/citología , Latencia del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA